SlideShare a Scribd company logo
Examples of Nutrition Claims
Claims about a popular diet that is supposed to change your
body, reverse a disease, or dramatically improve your health or
performance in some way.
Claims about a particular food, beverage or dietary supplement
that is supposed to help you lose weight, gain muscle, boost
immunity, improve mood or memory, lower blood cholesterol or
blood sugar levels, fight inflammation, remove toxins, prevent
or cure a disease, make your hair/nails/skin/digestion better,
slow aging…
Claims about a particular ingredient in foods/beverages that’s
supposed to be “bad,” “toxic,” or contribute to a particular
health problem (acne, autism, ADHD, PCOS, diabetes, cancer,
Alzheimer’s disease, aging, hormone disruption, infertility,
obesity, digestive problems)…
Sources of Nutrition Claims
Google!
Magazines, Newspapers, Blogs
Books, Videos, Documentaries
Advertisements, Social Media Influencers
Product label, brochure, website
Scientific peer-reviewed journals
How to choose a claim…
Examples:
Magazine article or blog claiming Intermittent fasting or whole
30 or keto is the answer to weight loss. Vitamin D/C/zinc and
COVID19.
LA Times article reporting on a new study that shows chocolate
or red wine protects the heart (in time for Valentine’s Day)
Book or Youtube video that claims sugar or wheat or gluten is
toxic
Documentary that claims plant-based diet best for performance
(The Game Changers)
Advertisement about new dietary supplement or “cleanse” for
brain health, skin health, digestive health (turmeric, collagen,
probiotics, spirulina, apple cider vinegar)
Website bodybuilding.com claiming need certain amount/type
of protein to get huge muscles. Or no soy/no dairy for PCOS or
fertility.
2
Evaluating Nutrition Research & Claims
Is the source credible & unbiased?
Author/credentials
“Nutritionists” vs. “Registered Dietitians” – what’s the
difference?
Self-proclaimed guru, fitness trainer, massage therapist, store
clerk
MDs, DCs, PhDs – are they always reliable?
Is there any conflict of interest? Are they trying to sell you
something?
Publication source
Internet site (.com or .org, .edu, .gov)
Magazine, newsletter, brochure, trade journal (paid advertising)
Peer-reviewed, professional/scientific journal
Most “nutritionists” have little to no formal education/degree
(e.g. famous people, fitness trainer/massage therapist/GNC or
health food store clerk). Some “nutritionists” do have a high
level of education/degree, but they may or may not be highly
educated in nutrition
Conflict of interest – Juice plus, herbal life, arbonne sales rep
directly trying to sell you something or
researcher/author/speaker could be employed/paid by the
company trying to sell something (funded by beef/dairy council)
Example of ephedra article in fitness magazine, local SCV
magazines
3
Evaluating Nutrition Research & Claims
How good is the research?
Study design
No systematic method at all
testimonials, anecdotal, before/after
Epidemiological / Observational Studies
only show correlations, NOT cause and effect (due to
confounding variables)
Intervention / Experimental Studies
CAN show cause & effect (because confounding variables are
controlled)
Best if study 1) randomized, 2) placebo-controlled, AND 3)
double-blinded
# of subjects / type of subjects
Study length
Especially when looking at long term weight loss and
health/safety
Epidemiological: Look at population of people’s lifestyle
habits and look for associations/correlations with health/disease
outcomes.
Ex. Study shows eating ice cream is associated with drowning
(that doesn’t show eating ice cream causes drowning…what
confounding factor might be the reason for the association?
People tend to eat more ice cream in the summer. People also
tend to swim more in the summer.)
Ex. Poor sperm quality linked to more phone/laptop use at
night. Confounding factor is less quality sleep linked with
both.
Ex: People who drink red wine have less heart disease (based on
Mediterranean diet, but could be less sat fat red meat, more
olive oil MUFAs, more fruits/vegs, more active lifestyle, and/or
genetics/ethnicity)
Ex: Americans eating more carbs and gaining weight (but
americans also eating more calories!)
Intervention: Give a group of people an intervention/treatment
and then see what effect it has on health/disease outcome.
Ex: Hydroxycut/diet pill 8 week placebo-controlled study with
same diet/exercise (but not randomized)
Caffeine revs metabolism initially but effects wear off, no
effect on weight/body fat loss. Long term keto diet effects?
Subjects: Animals/rats vs. humans., Post menopausal women
vs. young men (soy), congestive heart failure pts. Vs. young
men (arginine/nitric oxide supplements)
4
Be cautious with Observational Studies
Observational studies do NOT prove one thing causes the other.
They only show that two things are associated with one another.
The REAL cause may be due to a confounding variable that is
associated with both things.
NOTE: In this example, poor quality sleep is the confounding
variable since it results from late night electronics and is the
real cause of poor sperm quality.
Evaluating Nutrition Research & Claims
Are the findings put in perspective?
How many studies show a positive vs. negative or “null” effect?
Any warning about adverse side effects/risks?
Is the effective “dose” the same as what’s commonly consumed
in a food or supplement?
Any acknowledgement of other factors that have a more
significant impact on the outcome than the one being studied?
e.g. Risk of getting cancer from NOT eating vegetables is far
greater than the cancer risk from ingesting trace amounts of
pesticide residues on those vegetables.
# studies: Anyone can “cherry-pick” studies to find only those 2
that support the claim (without mentioning the hundreds that
don’t)
Safety: Keto and intermittent fasting in certain populations
harmful
Reasonable dose: omega 3s DHA added to foods
Physiologically significant: pesticide resides may be higher in
conventional than organic, but the cancer risk of NOT eating
any fruits and veggies is far greater
6
Using Library Resources
www.aging-us.com 2764 AGING
INTRODUCTION
Major depressive disorder (MDD) is viewed as a major
public health problem globally. MDD has a substantial
impact on society and individuals, such as increasing
economic burden and decreasing labor productivity
[1–3]. At a global level, more than 300 million people
are estimated to suffer from MDD, which is equivalent
to 4.4% of the world’s population [4]. However, the
pathogenesis of MDD is still unclear. Some theories
have been developed to explain the biological
mechanisms of MDD, such as neurotrophic alterations
www.aging-us.com AGING 2020, Vol. 12, No. 3
Research Paper
Age-specific differential changes on gut microbiota composition
in
patients with major depressive disorder
Jian-Jun Chen1,2,*,#, Sirong He3,*, Liang Fang2,4,*, Bin
Wang1, Shun-Jie Bai5, Jing Xie6, Chan-Juan
Zhou7, Wei Wang8, Peng Xie4,7,8,#
1Institute of Life Sciences, Chongqing Medical University,
Chongqing 400016, China
2Chongqing Key Laboratory of Cerebral Vascular Disease
Research, Chongqing Medical University, Chongqing
400016, China
3Department of Immunology, College of Basic Medicine,
Chongqing Medical University, Chongqing 400016, China
4Department of Neurology, Yongchuan Hospital of Chongqing
Medical University, Chongqing 402160, China
5Department of Laboratory, The First Affiliated Hospital of
Chongqing Medical University, Chongqing 400016, China
6Department of Endocrinology and Nephrology, Chongqing
University Central Hospital, Chongqing Emergency
Medical Center, Chongqing 400014, China
7NHC Key Laboratory of Diagnosis and Treatment on Brain
Functional Diseases, Chongqing Medical University,
Chongqing 400016, China
8Department of Neurology, The First Affiliated Hospital of
Chongqing Medical University, Chongqing 400016, China
*Equal contribution
#Co-senior authors
Correspondence to: Peng Xie, Jian-Jun Chen; email:
[email protected], [email protected]
Keywords: major depressive disorder, gut microbiota,
Firmicutes, Bacteroidetes, Actinobacteria
Received: November 21, 2019 Accepted: January 12, 2020
Published: February 10, 2020
Copyright: Chen et al. This is an open-access article distributed
under the terms of the Creative Commons Attribution License
(CC BY 3.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original author and
source are credited.
ABSTRACT
Emerging evidence has shown the age-related changes in gut
microbiota, but few studies were conducted to
explore the effects of age on the gut microbiota in patients with
major depressive disorder (MDD). This study was
performed to identify the age-specific differential gut
microbiota in MDD patients. In total, 70 MDD patients and 71
healthy controls (HCs) were recruited and divided into two
groups: young group (age 18-29 years) and middle-aged
group (age 30-59 years). The 16S rRNA gene sequences were
extracted from the collected fecal samples. Finally, we
found that the relative abundances of Firmicutes and
Bacteroidetes were significantly decreased and increased,
respectively, in young MDD patients as compared with young
HCs, and the relative abundances of Bacteroidetes
and Actinobacteria were significantly decreased and increased,
respectively, in middle-aged MDD patients as
compared with middle-aged HCs. Meanwhile, six and 25
differentially abundant bacterial taxa responsible for the
differences between MDD patients (young and middle-aged,
respectively) and their respective HCs were identified.
Our results demonstrated that there were age-specific
differential changes on gut microbiota composition in
patients with MDD. Our findings would provide a novel
perspective to uncover the pathogenesis underlying MDD.
mailto:[email protected]
mailto:[email protected]
www.aging-us.com 2765 AGING
and neurotransmission deficiency [5, 6]. However, none
of these theories has been universally accepted.
Therefore, there is a pressing need to identify novel
pathophysiologic mechanisms underlying this disease.
In recent years, mounting evidence has shown that gut
microbiota could play a vital role in every aspect of
physiology [7]. It is the largest and most direct external
environment of humans. Previous studies found that the
disturbance of gut microbiota had a crucial role in the
pathogenesis of many diseases [8–10]. Recent studies
reported that gut microbiota could affect the host brain
function and host behaviors through microbiota-gut-
brain axis [11, 12]. Using germ-free mice, we found that
gut microbiota could influence the gene levels in the
hippocampus of mice and lipid metabolism in the
prefrontal cortex of mice [13, 14]. Our clinical studies
demonstrated that the disturbance of gut microbiota
might be a contributory factor in the development of
MDD [15, 16].
Nowadays, emerging evidence has shown the age-
related changes in gut microbiota composition. For
example, Firmicutes is the dominant taxa during the
neonatal period, but Actinobacteria and Proteobacteria
are about to increase in three to six months [17]. While
in adults, Vemuri et al. reported that Bacteroidetes and
Firmicutes were the dominant taxa [18]. Meanwhile,
compared to younger individuals, the abundance of
Bacteroidetes is significantly higher in frailer older
individuals [19]. These results showed that there was a
close relationship between age and gut microbiota
composition. Ignoring this relationship would affect the
robust of results when exploring the mechanism of
action of gut microbiota in diseases. Therefore, to study
the relationship between gut microbiota and MDD
patients in different age groups, we recruited 52 young
subjects aged from 18 to 29 years (27 healthy controls
(HCs) and 25 MDD patients) and 89 middle-aged
subjects aged from 30 to 59 years (44 HCs and 45 MDD
patients). The main purpose of this study was to identify
the age-specific differential changes on gut microbiota
composition in MDD patients. Our results would
display the different changes of gut microbiota
composition along with age between HCs and MDD
patients.
RESULTS
Differential gut microbiota composition
As shown in Figure 1, the results of abundance-based
coverage estimator (ACE) and Chao1 showed that there
was no significant difference in OTU richness between
MDD patients (young and middle-aged, respectively)
and their respective HCs. However, the OPLS-DA
model built with young HCs and young MDD patients
showed an obvious difference in microbial abundances
between these two groups (Figure 2A). The relative
abundances of Firmicutes and Bacteroidetes were
Figure 1. Comparison of alpha diversity between HCs and MDD
patients. (A, B) ACE and Chao1 indexes showed no significant
differences between young HCs (n=27) and young MDD
patients (n=25); (C, D) ACE and Chao1 indexes showed no
significant differences
between middle-aged HCs (n=44) and middle-aged MDD
patients (n=45).
www.aging-us.com 2766 AGING
significantly decreased and increased, respectively, in
young MDD patients as compared with young HCs
(Figure 2B). Meanwhile, the OPLS-DA model built
with middle-aged HCs and middle-aged MDD patients
showed an obvious difference in microbial abundances
between these two groups (Figure 3A). The relative
abundances of Bacteroidetes and Actinobacteria were
significantly decreased and increased, respectively, in
middle-aged MDD patients as compared with middle-
aged HCs (Figure 3B).
Key discriminatory OTUs
In order to find out the gut microbiota primarily
responsible for the separation between MDD patients
(young and middle-aged, respectively) and their
respective HCs, the Random Forests classifier was used.
A total of 92 OTUs responsible for the separation
between young MDD patients and young HCs were
identified (Figure 4). These OTUs were mainly assigned
to the Families of Bacteroidaceae, Clostridiaceae_1,
Figure 2. 16S rRNA gene sequencing reveals changes to
microbial abundances in young MDD patients. (A) OPLS-DA
model
showed an obvious difference in microbial abundances between
the two groups (HCs, n=27; MDD, (n=25); (B) the relative
abundances of
Firmicutes and Bacteroidetes were significantly changed in
young MDD patients (n=25) as compared with young HCs
(n=27).
Figure 3. 16S rRNA gene sequencing reveals changes to
microbial abundances in middle-aged MDD patients. (A) OPLS-
DA
model showed an obvious difference in microbial abundances
between the two groups (HCs, n=44; MDD, (n=45); (B) the
relative abundances
of Bacteroidetes and Actinobacteria were significantly changed
in middle-aged MDD patients (n=45) as compared with middle-
aged HCs
(n=44).
www.aging-us.com 2767 AGING
Coriobacteriaceae, Erysipelotrichaceae, Lachnospiraceae,
Peptostreptococcaceae and Ruminococcaceae.
Meanwhile, a total of 122 OTUs responsible for the
separation between middle-aged MDD patients and
middle-aged HCs were identified (Figure 5). These OTUs
were mainly assigned to the Families of Lachnospiraceae,
Coriobacteriaceae, Streptococcaceae, Prevotellaceae,
Bacteroidaceae, Eubacteriaceae, Actinomycetaceae,
Sutterellaceae, Acidaminococcaceae, Erysipelotrichaceae,
Ruminococcaceae, and Porphyromonadaceae.
Figure 4. Heatmap of discriminative OTUs abundances between
young HCs (n=27) and young MDD patients (n=25).
Figure 5. Heatmap of discriminative OTUs abundances between
middle-aged HCs (n=44) and middle-aged MDD patients
(n=45).
www.aging-us.com 2768 AGING
Differentially abundant bacterial taxa
Differentially abundant bacterial taxa responsible for
the differences between MDD patients (young and
middle-aged, respectively) and their respective HCs
were identified by the metagenomic Linear
Discriminant Analysis (LDA) Effect Size (LEfSe)
approach (LDA score>2.0 and p-value<0.05). In total,
six bacterial taxa with statistically significant and
biologically consistent differences in young MDD
patients were identified (Figure 6). Meanwhile, fifteen
bacterial taxa with statistically significant and
biologically consistent differences in middle-aged MDD
patients were identified (Figure 7). In addition, using
Figure 6. Differentially abundant features identified by LEfSe
that characterize significant differences between young HCs
(n=27) and young MDD patients (n=25).
Figure 7. Differentially abundant features identified by LEfSe
that characterize significant differences between middle-aged
HCs (n=44) and middle-aged MDD patients (n=45).
www.aging-us.com 2769 AGING
the receiver operating characteristic (ROC) curve
analysis, we found that Clostridium_sensu_stricto,
Clostridium_XI and Clostridium_XVIII showed good
diagnostic performance (area under the curve (AUC)
>0.7) in diagnosing young MDD patients (Figure 8A–
8C). We also found that Anaerostipes, Streptococcus,
Blautia, Faecalibacterium and Roseburia showed good
diagnostic performance (AUC>0.7) in diagnosing
middle-aged MDD patients (Figure 8D–8H).
Effects of age on microbial abundances
Using the LEfSe approach, we identified four
differentially abundant bacterial taxa (the Family
level) between young HCs and middle-aged HCs
(Streptococcaceae, Coriobacteriaceae, Carnobacteriaceae
and Clostridiales_Incertae_Sedis_XIII) (Figure 9A);
we also identified six differentially abundant bacterial
taxa (the Family level) between young MDD patients
and middle-aged MDD patients (Prevotellaceae,
Acidaminococcaceae, Veillonellaceae Peptostrep-
tococcaceae, Lachnospiraceae and Ruminococcaceae)
(Figure 9B). Meanwhile, using the LEfSe approach, we
identified five differentially abundant bacterial taxa (the
Genus level) between young HCs and middle-aged HCs
(Streptococcus, Veillonella, Granulicatella, Collinsella
and Megamonas) (Figure 10A). All these bacterial
taxa were significantly decreased in middle-aged
HCs; we also identified nine differentially abundant
bacterial taxa (the Genus level) between young MDD
patients and middle-aged MDD patients (Megamonas,
Prevotella, Phascolarctobacterium, Anaerostipes,
Clostridium_XVIII, Gordonibacter, Eggerthella,
Clostridium_XI and Turicibacter) (Figure 10B).
Effects of medication on microbial abundances
To determinate the homogeneity of gut microbiota
composition between medicated and non-medicated
MDD patients, we firstly used the middle-aged HCs and
non-medicated middle-aged MDD patients to built
OPLS-DA model (Figure 11A). The results showed that
41 of the 44 middle-aged HCs and 30 of the 31 non-
medicated middle-aged MDD patients were correctly
diagnosed by the OPLS-DA model. Then, we used the
built model to predict class membership of 14
medicated middle-aged MDD patients. The T-predicted
scatter plot showed that 11 of the 14 medicated middle-
aged MDD patients were correctly predicted (Figure
11B). These finding indicated that the gut microbiota
composition of non-medicated middle-aged MDD
patients were distinct from middle-aged HCs, but not
from medicated middle-aged MDD patients.
DISCUSSION
Individuals in the different phases of life cycle (named
children, young, middle-aged and elderly) present
different biological characteristics and disease risks
[20]. Understanding the different characteristics of
patients in particular age phases could be facilitated to
prevent and treat diseases. According to the World
Health Organization reported, the prevalence rates of
depression vary by age, peaking in older adulthood. It
also occurs in children, but at a lower level compared
with older age groups. Here, we conducted this work to
investigate how the gut microbiota composition
changed in different age phases of MDD patients, and
found some age-specific differential gut microbiota in
Figure 8. Differential taxa (at the genus level) with AUC>0.7 in
diagnosing MDD patients from HCs. (A–C) the diagnostic
performances of three taxa in diagnosing young MDD patients
(n=25) from young HCs (n=27); (D–H) the diagnostic
performances of five taxa
in diagnosing middle-aged MDD patients (n=45) from middle-
aged HCs (n=44).
www.aging-us.com 2770 AGING
Figure 9. 16S rRNA gene sequencing reveals changes to
microbial abundances at family level (Mean±SEM). (A) the
abundances
of four taxonomic levels were significantly changed between
young HCs (n=27) and middle-aged HCs (n=44); (B) the
abundances of six
taxonomic levels were significantly changed between young
MDD patients (n=25) and middle-aged MDD patients (n=45).
Figure 10. 16S rRNA gene sequencing reveals changes to
microbial abundances at genus level (Mean±SEM). (A) the
abundances
of five taxonomic levels were significantly changed between
young HCs (n=27) and middle-aged HCs (n=44); (B) the
abundances of nine
taxonomic levels were significantly changed between young
MDD patients (n=25) and middle-aged MDD patients (n=45).
www.aging-us.com 2771 AGING
MDD patients. Our results could provide a new
perspective on exploring the pathogenesis of MDD.
Many previous studies focused on the effects of gut
microbiota on brain functions [21, 22]. However, few
studies have taken the effects of age on gut microbiota
into consideration when exploring the pathogenesis of
MDD. Our previous study found that the relative
abundances of Bacteroidetes and Actinobacteria were
significantly decreased and increased, respectively, in
MDD patients as compared with HCs [15]. But, in this
study, we found that the relative abundances of
Firmicutes and Bacteroidetes were significantly
decreased and increased, respectively, in young MDD
patients as compared with young HCs, and the relative
abundances of Bacteroidetes and Actinobacteria were
significantly decreased and increased, respectively, in
middle-aged MDD patients as compared with middle-
aged HCs. This disparity might be caused by the
different age structures. Meanwhile, only 35 key
discriminatory OTUs were significantly changed in both
young (92 key discriminatory OTUs) and middle-aged
(127 key discriminatory OTUs) MDD patients.
Moreover, the differentially abundant bacterial taxa in
young and middle-aged MDD patients were totally
different at both Family level and Genus level. These
results demonstrated that it was necessary to identify the
age-specific differential gut microbiota in patients with
MDD.
As far as we known, gut microbiota composition and its
function could be easily influenced by many factor,
such as gender, age, life experiences, dietary habit and
genetics. Mariat et al reported that the
Firmicutes/Bacteroidetes ratio of the human microbiota
could change with age [23]. Interestingly, here we
found that the relative abundance of Firmicutes was
significantly decreased in young MDD patients, but not
in middle-aged MDD patients; the relative abundance of
Bacteroidetes was significantly increased and
decreased, respectively, in young and middle-aged
MDD patients. In our previous studies, we did not
analyze the potential effects of medication on gut
microbiota composition in MDD patients [15, 16]. Here,
due to the small samples of young group, we only used
the middle-aged group to analyze the effects of
Figure 11. Assessment of gut microbiota composition in non-
medicated and medicated middle-aged MDD patients. (A)
middle-aged HCs (n=44) and non-medicated middle-aged MDD
patients (n=31) were effectively separated by the built OPLS-
DA model; (B) 14
medicated middle-aged MDD patients were correctly predicted
by the model.
www.aging-us.com 2772 AGING
medication on the gut microbiota composition. The
results showed that the medication seemed to have little
effects on gut microbiota composition in MDD patients.
However, our findings had to be cautiously interpreted
due to the relatively small samples using to analyze the
effects of medication on gut microbiota composition.
The relative abundance of genus Clostridium_XVIII
was not found to be significantly different between
MDD patients and HCs in our previous study [15].
However, in this study, we found that the relative
abundance of genus Clostridium_XVIII was
significantly decreased in young MDD patients
compared with young HCs, while increased in middle-
aged MDD patients compared with middle-aged HCs.
The reason of this disparity might be that age could
significantly affect the relative abundance of genus
Clostridium_XVIII in MDD patients, but not HCs: i)
compared to young MDD patients, the middle-aged
MDD patients had a significantly higher relative
abundance of genus Clostridium_XVIII; and ii) the
relative abundance of genus Clostridium_XVIII was
similar between young and middle-aged HCs.
Meanwhile, we found that the relative abundance of
genus Megamonas was significantly decreased in both
middle-aged HCs and middle-aged MDD patients
compared to their respective young populations. In
addition, most of differential bacterial taxa were
significantly decreased in middle-aged HCs compared
with young HCs, but only about half of differential
bacterial taxa were significantly decreased in middle-
aged MDD patients compared with young MDD
patients. Lozupone et al. reported that gut microbiota
could not only simply determine the certain host
characteristics, but also respond to signals from host via
multiple feedback loops [24]. Therefore, our results
suggested that age might have the different effects on
the gut microbiota composition of HCs and MDD
patients, and should always be considered in
investigating the relationship between MDD and gut
microbiota.
Limitations should be mentioned here. Firstly, the
number of HCs and MDD patients was relatively small,
and future works were still needed to further study and
support our results. Secondly, we only explored the age-
specific differential changes on gut microbiota
composition in patients with MDD; future studies
should further investigate the functions of these
identified differential gut microbiota using
metagenomic technology. Thirdly, all included subjects
were from the same site and ethnicity; thus, the
potential site- and ethnic-specific biases in microbial
phenotypes could not be ruled out, which might limit
the applicability of our results [25–28]. Fourthly, only
young and middle-aged groups were recruited, future
studies should recruit old-aged group and children
group to further identify the age-specific differential gut
microbiota in the different phases of life cycle. Fifthly,
we only investigated the differences in gut microbiota
between HCs and MDD patients on phylum level,
family level and genus level. Future studies were
needed to further explore the differences on other
levels, such as class level and species level. Sixthly, we
did not collect information on smoking, a factor which
could influence the gut microbiota composition. Future
studies were needed to analyze how the smoking
influenced the gut microbiota composition in the
different phases of life cycle of subjects. Finally, we
found that the medication status of subjects could not
significantly affect our results. However, limited by the
relatively small samples, this conclusion was needed
future studies to further validate.
In conclusion, in this study, we found that there were
age-specific differential changes on gut microbiota
composition in patients with MDD, and identified some
age-specific differentially abundant bacterial taxa in
MDD patients. Our findings would provide a novel
perspective to uncover the pathogenesis underlying
MDD, and potential gut-mediated therapies for MDD
patients. Limited by the small number of subjects, the
results of the present study were needed future studies
to validate and support.
MATERIALS AND METHODS
Subject recruitment
This study was approved by the Ethical Committee of
Chongqing Medical University and conformed to the
provisions of the Declaration of Helsinki. In total, there
were 27 young HCs (aged 18-29 years) and 25 young
MDD outpatients (aged 18-29 years) in the young
group; there were 44 middle-aged HCs (aged 30-59
years) and 45 middle-aged MDD outpatients (aged 30-
59 years) in the middle-aged group. Most of MDD
patients were first-episode drug-naïve depressed
subjects. There were only seven young MDD patients
and 14 middle-aged MDD patients receiving
medications. The detailed information of these included
subjects was described in Table 1. All HCs were
recruited from the Medical Examination Center of
Chongqing Medical University, and all MDD patients
were recruited from the psychiatric center of Chongqing
Medical University. MDD patients were screened in the
baseline interview by two experienced psychiatrists
using the DSM-IV (Diagnostic and Statistical Manual
of Mental Disorders, 4th Edition)-based Composite
International Diagnostic Interview (CIDI, version2.1).
The Hamilton Depression Rating Scale (HDRS) was
used to assess the depressive symptoms of each patient,
www.aging-us.com 2773 AGING
Table 1. Demographic and clinical characteristics of MDD
patients and HCsa.
Young group (18-29 years) Middle-aged group (30-59 years)
HC MDD p-value HC MDD p-value
Sample Size 27 25 – 44 45 –
Age (years)c 24.96±2.31 24.0±3.74 0.26 47.16±8.07 44.96±7.76
0.19
Sex (female/male) 19/8 18/7 0.89 34/10 31/14 0.37
BMI 21.53±2.37 22.13±2.24 0.35 23.23±2.33 22.64±2.64 0.26
Medication (Y/N) 0/27 7/18 – 0/44 14/31 –
HDRS scores 0.29±0.61 22.64±3.18 <0.00001 0.34±0.74
23.0±4.61 <0.00001
aAbbreviations: HDRS: Hamilton Depression Rating Scale;
HCs: healthy controls; MDD: major depressive disorder; BMI:
body
mass index.
and those patients with HDRS score >=17 were
included. Meanwhile, MDD patients were excluded if
they had other mental disorders, illicit drug use or
substance abuse, and were pregnant or menstrual
women. HCs were excluded if they were with mental
disorders, illicit drug use or systemic medical illness.
All the included subjects provided written informed
consent before sample collection.
16s rRNA gene sequencing
We used the standard PowerSoil kit protocol to extract
the bacterial genomic DNA from the fecal samples.
Briefly, we thawed the frozen fecal samples on ice and
pulverized the samples with a pestle and mortar in
liquid nitrogen. After adding MoBio lysis buffer into
the samples and mixing them, the suspensions were
centrifuged. The obtained supernatant was moved into
the MoBio Garnet bead tubes containing MoBio buffer.
Subsequently, we used the Roche 454 sequencing (454
Life Sciences Roche, Branford, PA, USA) to extract the
bacterial genomic DNA. The extracted V3-V5 regions
of 16S rRNA gene were polymerase chain reaction-
amplified with bar-coded universal primers containing
linker sequences for pyrosequencing [29].
The Mothur 1.31.2 (http://www.mothur.org/) was used
to quality-filtered the obtained raw sequences to
identify unique reads [30]. Raw sequences met any one
of the following criteria were excluded: i) less than
200bp or greater than 1000bp; ii) contained any
ambiguous bases, primer mismatches, or barcode
mismatches; and iii) homopolymer runs exceeding six
bases. The remaining sequences were assigned to
operational taxonomic units (OTUs) with 97%
threshold, and then taxonomically classified according
to Ribosomal Database Project (RDP) reference
database [31]. We used these taxonomies to construct
the summaries of the taxonomic distributions of OTUs,
and then calculated the relative abundances of gut
microbiota at different levels. The abovementioned
procedure and most of data were from our previous
studies [15, 16].
Statistical analysis
Richness was one of the two most commonly used alpha
diversity measurements. Here, we used two different
parameters (Chao1 and ACE) to estimate the OTU
richness [32, 33]. The orthogonal partial least squares
discriminant analysis (OPLS-DA) was a multivariate
method, which was used to remove extraneous variance
(unrelated to the group) from the sequencing datasets. The
LEfSe was a new analytical method for discovering the
metagenomic biomarker by class comparison. The
bacterial taxa with LDA score>2.0 were viewed as the
differentially abundant bacterial taxa responsible for the
differences between different groups. Here, both OPLS-
DA [34, 35] and LEfSe were used to reduce the
dimensionality of datasets and identify the differentially
abundant bacterial taxa (the Family level and Genus level)
that could be used to characterize the significant
differences between HCs and MDD patients. Meanwhile,
we used the Random Forest algorithm to identify the
critical discriminatory OTUs. The ROC curve analysis
was used to assess the diagnostic performance of these
identified differential bacterial taxa. The AUC was the
evaluation index. Finally, we used the LEfSe to reveal the
changes of microbial abundances at Family level and
Genus level in HCs and MDD patients, respectively.
ACKNOWLEDGMENTS
Our sincere gratitude is extended to Professors Delan
Yang and Hua Hu from Psychiatric Center of the First
Affiliated Hospital of Chongqing Medical University
for their efforts in sample collection.
CONFLICTS OF INTEREST
The authors declare no financial or other conflicts of
interest.
http://www.mothur.org/
www.aging-us.com 2774 AGING
FUNDING
This work was supported by the National Key R&D
Program of China (2017YFA0505700), the Non-profit
Central Research Institute Fund of Chinese Academy of
Medical Sciences (2019PT320002300), the Natural
Science Foundation Project of China (81820108015,
81701360, 81601208, 81601207), the Chongqing
Science and Technology Commission
(cstc2017jcyjAX0377), the Chongqing Yuzhong
District Science and Technology Commission
(20190115), and supported by the fund from the Joint
International Research Laboratory of Reproduction &
Development, Institute of Life Sciences, Chongqing
Medical University, Chongqing, China, and also
supported by the Scientific Research and Innovation
Experiment Project of Chongqing Medical University
(CXSY201862, CXSY201863).
REFERENCES
1. Yirmiya R, Rimmerman N, Reshef R. Depression as a
microglial disease. Trends Neurosci. 2015; 38:637–58.
https://doi.org/10.1016/j.tins.2015.08.001
PMID:26442697
2. Pan JX, Xia JJ, Deng FL, Liang WW, Wu J, Yin BM, Dong
MX, Chen JJ, Ye F, Wang HY, Zheng P, Xie P. Diagnosis
of major depressive disorder based on changes in
multiple plasma neurotransmitters: a targeted
metabolomics study. Transl Psychiatry. 2018; 8:130.
https://doi.org/10.1038/s41398-018-0183-x
PMID:29991685
3. Zhao H, Du H, Liu M, Gao S, Li N, Chao Y, Li R, Chen W,
Lou Z, Dong X. Integrative proteomics–metabolomics
strategy for pathological mechanism of vascular
depression mouse model. J Proteome Res. 2018;
17:656–69.
https://doi.org/10.1021/acs.jproteome.7b00724
PMID:29190102
4. Stringaris A. Editorial: what is depression? J Child
Psychol Psychiatry. 2017; 58:1287–89.
https://doi.org/10.1111/jcpp.12844 PMID:29148049
5. Luscher B, Shen Q, Sahir N. The GABAergic deficit
hypothesis of major depressive disorder. Mol
Psychiatry. 2011; 16:383–406.
https://doi.org/10.1038/mp.2010.120 PMID:21079608
6. Guilloux JP, Douillard-Guilloux G, Kota R, Wang X,
Gardier AM, Martinowich K, Tseng GC, Lewis DA, Sibille
E. Molecular evidence for BDNF- and GABA-related
dysfunctions in the amygdala of female subjects with
major depression. Mol Psychiatry. 2012; 17:1130–42.
https://doi.org/10.1038/mp.2011.113
PMID:21912391
7. Clemente JC, Ursell LK, Parfrey LW, Knight R. The
impact of the gut microbiota on human health: an
integrative view. Cell. 2012; 148:1258–70.
https://doi.org/10.1016/j.cell.2012.01.035
PMID:22424233
8. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ,
Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ,
Camporez JP, Shulman GI, Gordon JI, et al.
Inflammasome-mediated dysbiosis regulates
progression of NAFLD and obesity. Nature. 2012;
482:179–85.
https://doi.org/10.1038/nature10809 PMID:22297845
9. Peng W, Yi P, Yang J, Xu P, Wang Y, Zhang Z, Huang S,
Wang Z, Zhang C. Association of gut microbiota
composition and function with a senescence-
accelerated mouse model of Alzheimer’s Disease using
16S rRNA gene and metagenomic sequencing analysis.
Aging (Albany NY). 2018; 10:4054–65.
https://doi.org/10.18632/aging.101693
PMID:30562162
10. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L,
Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA,
Gordon JI, Chervonsky AV. Innate immunity and
intestinal microbiota in the development of Type 1
diabetes. Nature. 2008; 455:1109–13.
https://doi.org/10.1038/nature07336 PMID:18806780
11. Tillisch K. The effects of gut microbiota on CNS function
in humans. Gut Microbes. 2014; 5:404–10.
https://doi.org/10.4161/gmic.29232 PMID:24838095
12. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J,
Deng Y, Blennerhassett P, Macri J, McCoy KD, Verdu
EF, Collins SM. The intestinal microbiota affect central
levels of brain-derived neurotropic factor and behavior
in mice. Gastroenterology. 2011; 141:599–609,
609.e1–3.
https://doi.org/10.1053/j.gastro.2011.04.052
PMID:21683077
13. Chen JJ, Zeng BH, Li WW, Zhou CJ, Fan SH, Cheng K,
Zeng L, Zheng P, Fang L, Wei H, Xie P. Effects of gut
microbiota on the microRNA and mRNA expression in
the hippocampus of mice. Behav Brain Res. 2017;
322:34–41.
https://doi.org/10.1016/j.bbr.2017.01.021
PMID:28093256
14. Chen JJ, Xie J, Zeng BH, Li WW, Bai SJ, Zhou C, Chen W,
Wei H, Xie P. Absence of gut microbiota affects lipid
metabolism in the prefrontal cortex of mice. Neurol
Res. 2019; 41:1104–12.
https://doi.org/10.1080/01616412.2019.1675021
PMID:31587617
15. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L,
Chen J, Fan S, Du X, Zhang X, Yang D, Yang Y, et al. Gut
https://doi.org/10.1016/j.tins.2015.08.001
https://www.ncbi.nlm.nih.gov/pubmed/26442697
https://doi.org/10.1038/s41398-018-0183-x
https://www.ncbi.nlm.nih.gov/pubmed/29991685
https://doi.org/10.1021/acs.jproteome.7b00724
https://www.ncbi.nlm.nih.gov/pubmed/29190102
https://doi.org/10.1111/jcpp.12844
https://www.ncbi.nlm.nih.gov/pubmed/29148049
https://doi.org/10.1038/mp.2010.120
https://www.ncbi.nlm.nih.gov/pubmed/21079608
https://doi.org/10.1038/mp.2011.113
https://www.ncbi.nlm.nih.gov/pubmed/21912391
https://doi.org/10.1016/j.cell.2012.01.035
https://www.ncbi.nlm.nih.gov/pubmed/22424233
https://doi.org/10.1038/nature10809
https://www.ncbi.nlm.nih.gov/pubmed/22297845
https://doi.org/10.18632/aging.101693
https://www.ncbi.nlm.nih.gov/pubmed/30562162
https://doi.org/10.1038/nature07336
https://www.ncbi.nlm.nih.gov/pubmed/18806780
https://doi.org/10.4161/gmic.29232
https://www.ncbi.nlm.nih.gov/pubmed/24838095
https://doi.org/10.1053/j.gastro.2011.04.052
https://www.ncbi.nlm.nih.gov/pubmed/21683077
https://doi.org/10.1016/j.bbr.2017.01.021
https://www.ncbi.nlm.nih.gov/pubmed/28093256
https://doi.org/10.1080/01616412.2019.1675021
https://www.ncbi.nlm.nih.gov/pubmed/31587617
www.aging-us.com 2775 AGING
microbiome remodeling induces depressive-like
behaviors through a pathway mediated by the host’s
metabolism. Mol Psychiatry. 2016; 21:786–96.
https://doi.org/10.1038/mp.2016.44 PMID:27067014
16. Chen JJ, Zheng P, Liu YY, Zhong XG, Wang HY, Guo YJ,
Xie P. Sex differences in gut microbiota in patients with
major depressive disorder. Neuropsychiatr Dis Treat.
2018; 14:647–55.
https://doi.org/10.2147/NDT.S159322 PMID:29520144
17. Lim ES, Zhou Y, Zhao G, Bauer IK, Droit L, Ndao IM,
Warner BB, Tarr PI, Wang D, Holtz LR. Early life
dynamics of the human gut virome and bacterial
microbiome in infants. Nat Med. 2015; 21:1228–34.
https://doi.org/10.1038/nm.3950 PMID:26366711
18. Vemuri R, Gundamaraju R, Shastri MD, Shukla SD,
Kalpurath K, Ball M, Tristram S, Shankar EM, Ahuja K,
Eri R. Gut Microbial Changes, Interactions, and Their
Implications on Human Lifecycle: An Ageing
Perspective. Biomed Res Int. 2018; 2018:4178607.
https://doi.org/10.1155/2018/4178607
PMID:29682542
19. Claesson MJ, Cusack S, O’Sullivan O, Greene-Diniz R, de
Weerd H, Flannery E, Marchesi JR, Falush D, Dinan T,
Fitzgerald G, Stanton C, van Sinderen D, O’Connor M,
et al. Composition, variability, and temporal stability of
the intestinal microbiota of the elderly. Proc Natl Acad
Sci USA. 2011 (Suppl 1); 108:4586–91.
https://doi.org/10.1073/pnas.1000097107
PMID:20571116
20. Chia CW, Egan JM, Ferrucci L. Age-Related Changes in
Glucose Metabolism, Hyperglycemia, and
Cardiovascular Risk. Circ Res. 2018; 123:886–904.
https://doi.org/10.1161/CIRCRESAHA.118.312806
PMID:30355075
21. Cryan JF, Dinan TG. Mind-altering microorganisms: the
impact of the gut microbiota on brain and behaviour.
Nat Rev Neurosci. 2012; 13:701–12.
https://doi.org/10.1038/nrn3346
PMID:22968153
22. Gareau MG, Wine E, Rodrigues DM, Cho JH, Whary MT,
Philpott DJ, Macqueen G, Sherman PM. Bacterial
infection causes stress-induced memory dysfunction in
mice. Gut. 2011; 60:307–17.
https://doi.org/10.1136/gut.2009.202515
PMID:20966022
23. Mariat D, Firmesse O, Levenez F, Guimarăes V, Sokol H,
Doré J, Corthier G, Furet JP. The
Firmicutes/Bacteroidetes ratio of the human
microbiota changes with age. BMC Microbiol. 2009;
9:123.
https://doi.org/10.1186/1471-2180-9-123
PMID:19508720
24. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK,
Knight R. Diversity, stability and resilience of the
human gut microbiota. Nature. 2012; 489:220–30.
https://doi.org/10.1038/nature11550 PMID:22972295
25. Chen JJ, Bai SJ, Li WW, Zhou CJ, Zheng P, Fang L, Wang
HY, Liu YY, Xie P. Urinary biomarker panel for
diagnosing patients with depression and anxiety
disorders. Transl Psychiatry. 2018; 8:192.
https://doi.org/10.1038/s41398-018-0245-0
PMID:30232320
26. Hou L, Wei X, Zhuo Y, Qin L, Yang F, Zhang L, Song X.
GC-MS-based metabolomics approach to diagnose
depression in hepatitis B virus-infected patients with
middle or old age. Aging (Albany NY). 2018;
10:2252–65.
https://doi.org/10.18632/aging.101535
PMID:30178754
27. Chen JJ, Xie J, Zeng L, Zhou CJ, Zheng P, Xie P. Urinary
metabolite signature in bipolar disorder patients
during depressive episode. Aging (Albany NY). 2019;
11:1008–18.
https://doi.org/10.18632/aging.101805
PMID:30721880
28. Chen JJ, Xie J, Li WW, Bai SJ, Wang W, Zheng P, Xie P.
Age-specific urinary metabolite signatures and
functions in patients with major depressive disorder.
Aging (Albany NY). 2019; 11:6626–37.
https://doi.org/10.18632/aging.102133
PMID:31493765
29. Tamaki H, Wright CL, Li X, Lin Q, Hwang C, Wang S,
Thimmapuram J, Kamagata Y, Liu WT. Analysis of 16S
rRNA amplicon sequencing options on the Roche/454
next-generation titanium sequencing platform. PLoS
One. 2011; 6:e25263.
https://doi.org/10.1371/journal.pone.002 5263
PMID:21966473
30. Yang S, Liebner S, Alawi M, Ebenhöh O, Wagner D.
Taxonomic database and cut-off value for processing
mcrA gene 454 pyrosequencing data by MOTHUR. J
Microbiol Methods. 2014; 103:3–5.
https://doi.org/10.1016/j.mimet.2014.05. 006
PMID:24858450
31. Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y,
Brown CT, Porras-Alfaro A, Kuske CR, Tiedje JM.
Ribosomal Database Project: data and tools for high
throughput rRNA analysis. Nucleic Acids Res. 2014;
42:D633–42.
https://doi.org/10.1093/nar/gkt1244 PMID:24288368
32. Countway PD, Gast RJ, Dennett MR, Savai P, Rose JM,
Caron DA. Distinct protistan assemblages characterize
the euphotic zone and deep sea (2500 m) of the
western North Atlantic (Sargasso Sea and Gulf Stream).
https://doi.org/10.1038/mp.2016.44
https://www.ncbi.nlm.nih.gov/pubmed/27067014
https://doi.org/10.2147/NDT.S159322
https://www.ncbi.nlm.nih.gov/pubmed/29520144
https://doi.org/10.1038/nm.3950
https://www.ncbi.nlm.nih.gov/pubmed/26366711
https://doi.org/10.1155/2018/4178607
https://www.ncbi.nlm.nih.gov/pubmed/29682542
https://doi.org/10.1073/pnas.1000097107
https://www.ncbi.nlm.nih.gov/pubmed/20571116
https://doi.org/10.1161/CIRCRESAHA.118.312806
https://www.ncbi.nlm.nih.gov/pubmed/30355075
https://doi.org/10.1038/nrn3346
https://www.ncbi.nlm.nih.gov/pubmed/22968153
https://doi.org/10.1136/gut.2009.202515
https://www.ncbi.nlm.nih.gov/pubmed/20966022
https://doi.org/10.1186/1471-2180-9-123
https://www.ncbi.nlm.nih.gov/pubmed/19508720
https://doi.org/10.1038/nature11550
https://www.ncbi.nlm.nih.gov/pubmed/22972295
https://doi.org/10.1038/s41398-018-0245-0
https://www.ncbi.nlm.nih.gov/pubmed/30232320
https://doi.org/10.18632/aging.101535
https://www.ncbi.nlm.nih.gov/pubmed/30178754
https://doi.org/10.18632/aging.101805
https://www.ncbi.nlm.nih.gov/pubmed/30721880
https://doi.org/10.18632/aging.102133
https://www.ncbi.nlm.nih.gov/pubmed/31493765
https://doi.org/10.1371/journal.pone.0025263
https://www.ncbi.nlm.nih.gov/pubmed/21966473
https://doi.org/10.1016/j.mimet.2014.05.006
https://www.ncbi.nlm.nih.gov/pubmed/24858450
https://doi.org/10.1093/nar/gkt1244
https://www.ncbi.nlm.nih.gov/pubmed/24288368
www.aging-us.com 2776 AGING
Environ Microbiol. 2007; 9:1219–32.
https://doi.org/10.1111/j.1462-2920.2007.01243.x
PMID:17472636
33. Zuendorf A, Bunge J, Behnke A, Barger KJ, Stoeck T.
Diversity estimates of microeukaryotes below the
chemocline of the anoxic Mariager Fjord, Denmark.
FEMS Microbiol Ecol. 2006; 58:476–91.
https://doi.org/10.1111/j.1574-6941.2006.00171.x
PMID:17117990
34. Shankar V, Agans R, Holmes B, Raymer M, Paliy O. Do
gut microbial communities differ in pediatric IBS and
health? Gut Microbes. 2013; 4:347–52.
https://doi.org/10.4161/gmic.24827
PMID:23674073
35. Ramadan Z, Xu H, Laflamme D, Czarnecki-Maulden G,
Li QJ, Labuda J, Bourqui B. Fecal microbiota of cats with
naturally occurring chronic diarrhea assessed using 16S
rRNA gene 454-pyrosequencing before and after
dietary treatment. J Vet Intern Med. 2014; 28:59–65.
https://doi.org/10.1111/jvim.12261
PMID:24592406
https://doi.org/10.1111/j.1462-2920.2007.01243.x
https://www.ncbi.nlm.nih.gov/pubmed/17472636
https://doi.org/10.1111/j.1574-6941.2006.00171.x
https://www.ncbi.nlm.nih.gov/pubmed/17117990
https://doi.org/10.4161/gmic.24827
https://www.ncbi.nlm.nih.gov/pubmed/23674073
https://doi.org/10.1111/jvim.12261
https://www.ncbi.nlm.nih.gov/pubmed/24592406
RESEARCH Open Access
Associations between gut microbiota and
Alzheimer’s disease, major depressive
disorder, and schizophrenia
Zhenhuang Zhuang1, Ruotong Yang1, Wenxiu Wang1, Lu
Qi2,3* and Tao Huang1,4,5,6*
Abstract
Background: Growing evidence has shown that alterations in the
gut microbiota composition were associated
with a variety of neuropsychiatric conditions. However, whether
such associations reflect causality remains
unknown. We aimed to reveal the causal relationships among
gut microbiota, metabolites, and neuropsychiatric
disorders including Alzheimer’s disease (AD), major depressive
disorder (MDD), and schizophrenia (SCZ).
Methods: A two-sample bi-directional Mendelian randomization
analysis was performed by using genetic variants from
genome-wide association studies as instrumental variables for
gut microbiota, metabolites, AD, MDD, and SCZ, respectively.
Results: We found suggestive associations of host-genetic-
driven increase in Blautia (OR, 0.88; 95%CI, 0.79–0.99;
P = 0.028) and elevated γ-aminobutyric acid (GABA) (0.96;
0.92–1.00; P = 0.034), a downstream product of
Blautia-dependent arginine metabolism, with a lower risk of
AD. Genetically increased Enterobacteriaceae family
and Enterobacteriales order were potentially associated with a
higher risk of SCZ (1.09; 1.00–1.18; P = 0.048),
while Gammaproteobacteria class (0.90; 0.83–0.98; P = 0.011)
was related to a lower risk for SCZ. Gut
production of serotonin was potentially associated with an
increased risk of SCZ (1.07; 1.00–1.15; P = 0.047).
Furthermore, genetically increased Bacilli class was related to a
higher risk of MDD (1.07; 1.02–1.12; P = 0.010).
In the other direction, neuropsychiatric disorders altered gut
microbiota composition.
Conclusions: These data for the first time provide evidence of
potential causal links between gut microbiome
and AD, MDD, and SCZ. GABA and serotonin may play an
important role in gut microbiota-host crosstalk in
AD and SCZ, respectively. Further investigations in
understanding the underlying mechanisms of associations
between gut microbiota and AD, MDD, and SCZ are required.
Keywords: Gut microbiota, Neuropsychiatric disorder,
Mendelian randomization, Genetic association, Causality
Background
The human intestine comprises a very complex group of
gut microbiota, which influence the risk of neuropsychiatric
disorders [1, 2]. Accumulating evidence has suggested that
microbiota metabolites such as neurotransmitters and
short-chain fatty acids (SCFAs) may play a central role in
microbiota-host crosstalk that regulates the brain function
and behavior [3, 4]. Therefore, to understand the mechan-
ism of the gut-brain axis in neuropsychiatric disorders may
have clinical benefits.
Observational studies, most of case-control designs, have
shown differences in the composition of the gut microbiota
between healthy individuals and patients with neuropsychi-
atric disorders such as Alzheimer’s disease (AD), major
depression disorder (MDD), and schizophrenia (SCZ);
© The Author(s). 2020 Open Access This article is licensed
under a Creative Commons Attribution 4.0 International
License,
which permits use, sharing, adaptation, distribution and
reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if
changes were made. The images or other third party material in
this article are included in the article's Creative Commons
licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article's Creative
Commons
licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder. To view a copy
of this licence, visit
http://creativecommons.org/licenses/by/4.0/.
The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to
the
data made available in this article, unless otherwise stated in a
credit line to the data.
* Correspondence: [email protected]; [email protected]
2Department of Epidemiology, School of Public Health and
Tropical
Medicine, Tulane University, New Orleans, LA, USA
1Department of Epidemiology & Biostatistics, School of Public
Health, Peking
University, 38 Xueyuan Road, Beijing 100191, China
Full list of author information is available at the end of the
article
Zhuang et al. Journal of Neuroinflammation (2020)
17:288
https://doi.org/10.1186/s12974-020-01961-8
http://crossmark.crossref.org/dialog/?doi=10.1186/s12974-020-
01961-8&domain=pdf
http://orcid.org/0000-0002-0328-1368
http://creativecommons.org/licenses/by/4.0/
http://creativecommons.org/publicdomain/zero/1.0/
mailto:[email protected]
mailto:[email protected]
however, such associations substantially differed across
studies [5–7]. Noteworthy, genome-based metabolic mod-
eling of the human gut microbiota revealed that several
genera have predictive capability to produce or consume
neurotransmitters (called microbial neurotransmitters) such
as γ-aminobutyric acid (GABA) and serotonin [8, 9], which
have been consistently shown to played a key role in the
regulation of brain function [10, 11]. A meta-analysis of 35
observational studies reported that increased GABA levels
were associated with a lower risk of AD [12]. In addition, a
previous study (n = 40) reported that plasma serotonin was
lower and platelet serotonin was higher in SCZ patients
compared with controls [13], while another study showed
that lower platelet serotonin concentrations were associated
with depressive symptoms of SCZ (n = 364) [14]. There is
no doubt that these small observational studies were sus-
ceptible to confounding bias and reverse causation. It is
crucial to elucidate whether such associations reflect causal
relations or spurious correlations due to bias.
Mendelian randomization (MR), which overcomes the
bias due to confounding and reverse causation above-
mentioned, has been widely used to assess causal rela-
tionships by exploiting genetic variants as instrumental
variables of the exposure [15]. Recent genetic studies
have demonstrated that the host genetic variants influ-
ence the gut microbiota composition [16–18]. Thus,
such findings allowed us to deploy an MR approach to
infer the mutually causal relations of gut microbiota and
metabolites with neuropsychiatric disorders.
Therefore, we for the first time applied a two-sample
bi-directional MR approach to detect causal relation-
ships among gut microbiota, metabolites, and diverse
forms of neuropsychiatric disorders including AD, SCZ,
and MDD.
Methods
Study design overview
We employed a two-sample bi-directional MR approach
to investigate the causal relationships among gut micro-
biota, metabolites, and AD, MDD, or SCZ using
summary-level data from large genome-wide association
studies (GWASs) for gut microbiota and AD, MDD, or
SCZ. Ethical approval for each study included in the MR
analysis can be found in the original articles [19–23].
Data sources and instruments
Gut microbiota
We leveraged summary statistics from a GWAS of gut
microbiota conducted among two independent but geo-
graphically matched cohorts of European ancestry (n =
1812) using 16S rRNA gene sequencing (Table 1) [19],
which yielded a total of 38 and 374 identified phyla and
genera respectively. The GWAS defined a “core measur-
able microbiota” after removing rare bacteria and
investigating associations between host genetic variants
and specific bacterial traits, including 40 operational
taxonomic units (OTUs) and 58 taxa ranging from the
genus to the phylum level. Accordingly, the GWAS fur-
ther identified 54 genome-wide significant associations
involving 40 loci and 22 bacterial traits (meta-analysis P
< 5 × 10−8). We selected single nucleotide polymor-
phisms (SNPs) at thresholds for genome-wide signifi-
cance (P < 5 × 10−8) from this GWASs as genetic
instruments (Table S1).
Gut microbial metabolites
Considering the important roles of gut microbiota-
derived metabolites in microbiota-host crosstalk in the
brain function and behavior, we further chose key me-
tabolites with available GWAS, including propionic acid,
β-hydroxybutyric acid (BHB), serotonin, GABA, tri-
methylamine N-oxide (TMAO), betaine, choline, and
carnitine. These gut microbial metabolites play crucial
roles in maintaining a healthy neuropsychiatric function,
and if dysregulated, potentially causally linked to neuro-
psychiatric disorders according to previous studies [3,
24, 25]. We searched PubMed for GWASs of the gut
metabolites and leveraged summary-level data from a re-
cent GWAS of the human metabolome conducted
among 2076 participants of the Framingham Heart
Study (Table 1) [20]. Since few loci identified by gut me-
tabolite GWAS have reached the level of genome-wide
significance, we only selected SNPs at thresholds for
suggestive genome-wide significance (P < 1 × 10−5) from
the GWAS for each metabolite (Table S2).
Neuropsychiatric disorders
We searched PubMed for GWASs of the neuropsychi-
atric disorders and identified SNPs with genome-wide
significant (P < 5 × 10−8) associations for AD [21], MDD
[22], and SCZ [23], respectively (Table 1, Table S3).
Summarized data for AD were obtained from the Inter-
national Genomics of Alzheimer’s Project (IGAP), in-
cluding 25,580 AD cases and 48,466 controls, and the
analysis was adjusted for age, sex, and principal compo-
nents when necessary [21]. Genetic associations for
MDD were obtained from Psychiatric Genomics Consor-
tium 29 (PGC29) including135,458 MDD cases and 344,
901 controls, using sex and age as covariates [22]. Gen-
etic associations for SCZ were obtained from a meta-
analysis of Sweden and PGC including 13,833 SCZ cases
and 18,310 controls [23]. Detailed information on diag-
nostic criteria for AD, MDD, and SCZ are provided in
Table S4. These GWASs identified 19 SNPs for AD, 44
SNPs for MDD, and 24 SNPs for SCZ (P < 5 × 10−8), re-
spectively (Table S3).
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 2 of 9
Statistical analysis
For instrumental variables, we only selected independent
genetic variants which are not in linkage disequilibrium
(LD) (defined as r2 < 0.1) with other genetic variants
based on European ancestry reference data from the
1000 Genomes Project. We chose the variant with the
lowest P value for association with the exposure when
genetic variants were in LD. Moreover, for SNPs that
were not available in GWASs of the outcome, we used
the LD proxy search on the online platform (https://
snipa.helmholtz-muenchen.de/snipa3/index.php/) to re-
place them with the proxy SNPs identified in high-LD
(r2 > 0.8) or discard them if the proxies were not avail -
able. Power calculations for the MR study were con-
ducted based on the website: mRnd (http://cnsgenomics.
com/shiny/mRnd/).
We combined MR estimates by using inverse variance
weighting (IVW) as primary method. Weighted mode,
weighted median, and MR-Egger methods were used as
sensitivity analyses. Detailed information about the MR
methods mentioned above has been explained previously
[26, 27]. The MR-Egger method examined for unknown
horizontal pleiotropy as indicated by a non-zero inter-
cept value. We also applied leave-one-SNP-out approach
assessing the effects of removing these SNPs from the
MR analysis to rule out potential pleiotropic effects. Ef-
fect estimates are reported in beta values for the con-
tinuous outcome and ORs (95% CIs) for binary
outcome. Bonferroni correction was used to adjust for
multiple comparisons, giving a cutoff of P = 7.6 × 10−4
for the causal effect of gut microbiota on disorders and a
cutoff of P = 1.7 × 10−4 for reverse causation.
The MR analyses were conducted in the R version
3.5.1 computing environment (http://www.r-project.org)
using the TwoSampleMR package (R project for Statis-
tical Computing). This package harmonized effect of the
exposure and outcome data sets including combined in-
formation on SNPs, including phenotypes, effect alleles,
effect allele frequencies, effect sizes, and standard errors
for each SNP. In addition, we assumed that all alleles are
presented on the forward strand in harmonization. In
conclusion, the bi-directional MR results using the full
set of selected SNPs.
Results
Associations of gut microbiota and metabolites with
neuropsychiatric disorders
We found suggestive evidence of a protective effect of
the host-genetic-driven increase in Blautia on the risk of
AD (per relative abundance: OR, 0.88; 95% CI, 0.79–
0.99; P = 0.028) (Fig. 1, Figure S1). Importantly, we fur-
ther observed suggestive evidence that genetically ele-
vated gut metabolite GABA was associated with a lower
risk of AD (per 10 units: 0.96; 0.92–1.00; P = 0.034)
(Figs. 1 and 2).
Furthermore, the host-genetic-driven increases in En-
terobacteriaceae family and Enterobacteriales order were
potentially related to a higher risk of SCZ (1.09; 1.00–
1.18; P = 0.048), while Gammaproteobacteria class was
related to a lower risk of SCZ (0.90; 0.83–0.98; P =
0.011) (Fig. 1, Figure S1). Interestingly, gut production of
serotonin was potentially associated with a higher risk of
SCZ (1.07; 1.00–1.15; P = 0.047) (Figs. 1 and 3). In
addition, we found suggestive association of the host-
genetic-driven increase in Bacilli class with a higher risk
of MDD (1.07; 1.02–1.12; P = 0.010) (Fig. 1, Figure S1).
Sensitivity analysis yielded similar results for the causal
effects of gut microbiota on neuropsychiatric disorders, and
no horizontal pleiotropy or outliers were observed (Tables
S5 and S6). No significant results were found for any of
other selected gut microbiota or metabolites with neuro-
psychiatric disorders (Table S7). MR power calculation
showed strong power to detect significant (P < 7.6 × 10−4)
causal effect (OR = 1.2) for most of gut microbiota with the
risk of AD, MDD, and SCZ, respectively (Table S8).
Associations of neuropsychiatric disorders with gut
microbiota
In the opposite direction, we applied the MR method to
investigate the causal relationship of neuropsychiatric
Table 1 Description of gut microbiota, metabolites, and
neuropsychiatric disorders
Traits Consortium or study Sample size Populations Journal
Year
Gut
Gut microbiota PopGen/FoCus 1812 individuals European Nat
Genet. 2016
Gut metabolites FHS 2076 individuals European Cell Metab.
2013
Neuropsychiatric disorders
Alzheimer’s disease IGAPa 25,580 cases and 48,466 controls
European Nat Genet. 2013
Major depression disorder
PGC29/deCODE/GenScotland/GERA/iPSYCH/UK
Biobank/23andMeD
135,458 cases and 344,901 controls European Nat Genet. 2018
Schizophrenia Sweden/PGC 21,246 cases and 38,072 controls
European Nat Genet. 2013
FoCus Food-Chain Plus, GERA Genetic Epidemiology Research
on Adult Health and Aging, PGC Psychiatric Genomics
Consortium
a IGAP includes the Alzheimer’s Disease Genetics Consortium
(ADGC), the Cohorts for Heart and Aging Research in Genomic
Epidemiology consortium (CHARGE),
the European Alzheimer’s disease Initiative (EADI), and the
Genetic and Environmental Risk in Alzheimer’s disease
consortium (GERAD)
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 3 of 9
https://snipa.helmholtz-muenchen.de/snipa3/index.php
https://snipa.helmholtz-muenchen.de/snipa3/index.php
http://cnsgenomics.com/shiny/mRnd/
http://cnsgenomics.com/shiny/mRnd/
http://www.r-project.org
disorders with gut microbiota. We found a suggestive as-
sociation of AD with lower relative abundance of Erysi -
pelotrichaceae family, Erysipelotrichales order, and
Erysipelotrichia class (per 1-unit odds ratio: Beta±SE, −
0.274 ± 0.090; P = 0.003) and higher relative abundance
of unclassified Porphyromonadaceae (0.351 ± 0.170; P =
0.040) (Fig. 1, Table S9). Additionally, MDD was associ -
ated with higher relative abundance of unclassified Clos-
tridiales (0.577 ± 0.241; P = 0.017), OTU16802
Bacteroides (0.842 ± 0.386; P = 0.029), and unclassified
Prevotellaceae (0.978 ± 0.464; P = 0.035) (Fig. 1, Table
S9). We further identified that SCZ was nominally re-
lated to 2 genera, including higher relative abundance of
OTU10589 unclassified Enterobacteriaceae (0.457 ±
0.220; P = 0.037) and lower relative abundance of un-
classified Erysipelotrichaceae (− 0.248 ± -0.019; P =
0.045) (Fig. 1, Table S9).
Associations were almost consistent in sensitivity ana-
lyses using the weighted mode and weighted median
methods. The MR-Egger method showed directional
pleiotropy in the analysis of association between MDD
and OTU16802 Bacteroides (P = 0.022) but not in any
other potential significant associations (Table S9). How-
ever, we had limited power (all less than 50%) to test sig-
nificant (P < 1.7 × 10−4) causal effect (Beta = 0.5) of the
risk of AD, MDD, and SCZ on specific gut microbiota
(data not shown), possibly due to small sample size of
the gut microbiota GWAS.
Discussion
In this two-sample bi-directional MR study, we found
suggestive evidence of causal relationships of Blautia
with AD, of Enterobacteriaceae family, Enterobacteriales
order, and Gammaproteobacteria class with SCZ, and of
Bacilli class with MDD. More importantly, several neu-
rotransmitters such as GABA and serotonin produced
by gut microbiota were also potentially linked to the
risks of neuropsychiatric disorders, implying their im-
portant roles in microbiota-host crosstalk in the brain
function and behavior. In the other direction, our results
suggested that neuropsychiatric disorders, including AD,
SCZ, and MDD might alter the composition of gut
microbiota.
Microbiota-gut-brain communication has been shown
to play a key role in cognitive function [2]. However,
animal studies regarding the effects of Blautia genus on
AD have yielded conflicting results, but extrapolating
these findings to human beings is challenging [28, 29]. A
cohort study (n = 108) reported that decreased propor-
tion of Blautia hansenii was associated with a higher
risk of AD [30], while two case-control studies observed
that Blautia were more abundant in AD patients [5, 31].
Fig. 1 Schematic representation of the present study,
highlighting for each step of the study design and the
significant results obtained. We
aimed to estimate causal relationships between gut microbiota
(98 individual bacterial traits) and neuropsychiatric disorders
(Alzheimer’s disease,
major depression disorder, and schizophrenia) using a bi-
directional Mendelian randomization (MR) approach (step 1).
Then, we performed a two-
sample MR analysis to identify which microbiota metabolites
associated with these disorders (step 2). Finally, we identified
14 individual bacterial
traits and 2 gut metabolites to be associated with these
disorders. GABA, γ-aminobutyric acid; SCFA, short-chain fatty
acids
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 4 of 9
Although the direction of associations between Blautia
and the risk of AD substantially differed across studies,
one consistent finding was that gut microbial neuro-
transmitter GABA, a downstream product of Blautia-
dependent arginine metabolism, was related to a reduced
risk of AD. Notably, lower levels of gut product of
GABA were observed in patients with AD in several
case-control studies [32, 33]. In this bi-directional MR
study, our results for the first time provide evidence of a
causal relationship between relative abundance of Blau-
tia and AD. More importantly, we demonstrated that el-
evated GABA was potentially associated with a lower
risk of AD. Our findings supported previous meta-
analysis of 35 observational studies which suggested that
GABA level in AD were significantly lower than that of
controls [12]. Our findings suggest that GABA produced
by gut microbiota may play an important role in
microbiota-host crosstalk in the brain function and be-
havior. Although not significant, our findings show very
similar association directions for Blautia with MDD and
SCZ. Our findings are in line with recent studies which
indicated that decreased Blautia was associated with an
increased risk of autistic spectrum disorder (ASD), sug-
gesting a general change associated with psychiatric dis-
orders [34].
There are many potential pathways linking specific gut
microbiota to AD, among which metabolites produced
by gut microbiota may play an important role. It is
worth noting that GABA, as a primary inhibitory neuro-
transmitter in the human central nervous system (CNS),
has been shown to shape neurological processes and
cognition [35]. Recent evidence has demonstrated that
GABAergic functions could be an essential factor in the
whole stage of AD pathogenesis which seemed to be
more resistant to neurodegenerative changes in aged
brain [36, 37]. Our MR results that increased GABA
levels was potentially associated with a lower risk of AD
lent further support to the hypotheses. The biological
mechanisms of GABA production include degradation
of putrescine, decarboxylation of glutamate, or from ar-
ginine or ornithine [8]. Interestingly, the genus Blautia
has shown a strong correlation with arginine metabolism
[38], which may be involved in AD pathogenesis by
regulating its downstream products such as GABA, sup-
porting the potential pathway [39]. Since AD does not
break out suddenly but develops through a long pro-
dromal phase instead, it is plausible that our findings may
be potentially effective in early interventions of such dis -
order in the future by targeting the microbiota (e.g., gut
microbiota transplantation, psychobiotics, or antibiotics).
Fig. 2 Causal effect of GABA with the risk of AD. a Schematic
representation of the MR analysis results: genetically
determined higher GABA
plasma levels were potentially associated with a lower risk of
AD. b The odds ratios (95% confidence interval) for AD per 10
units increase in
GABA, as estimated in the inverse-variance weighted, weighted
mode, weighted median, and MR-Egger MR analysis. The
intercept of MR-Egger
can be interpreted as a test of overall unbalanced horizontal
pleiotropy. c The scatter plot represents instruments association
including AD
associations (y-axis) against instrument GABA associations (x-
axis). The tunnel plot represents instrument precision (i.e.,
instrument AD regression
coefficients divided by the correspondent instrument GABA
SEs) (y-axis) against individual instrument ratio estimates in
log odds ratio of AD (x-
axis). βIV indicates odds ratio estimate per 1-ln 10 units
increment in GABA levels. AD, Alzheimer’s disease; OR, odds
ratio; CI, confidence interval;
SNP, single-nucleotide polymorphism; SE, standard error; IVW,
inverse variance weighted
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 5 of 9
Recently, Enterobacteriales family and Gammaproteo-
bacteria class have been identified to be important bio-
markers of SCZ in recent cross-sectional studies,
consistent with our findings [6, 40]. Furthermore, a case-
control study (n = 364) identified a strong relationship
of lower platelet serotonin concentrations with depres-
sive symptoms of SCZ [14]. However, available evidence
is still largely inadequate since observational studies
mainly rely on self-reported information and are suscep-
tible to confounding (e.g., diet and health status) and re-
verse causation bias. Ertugrul et al. observed plasma
serotonin increased while platelet serotonin decreased in
SCZ patients after clinical treatments, which was incon-
sistent with our findings [13]. In addition, our results
support the finding that increased Bacilli is potentially
associated with a higher risk of MDD, possibly involving
dopamine metabolism which might play a role in the
major symptoms of MDD [41, 42]. A meta-analysis of
RCTs showed that probiotics, typically including Lacto-
bacillus and Bifidobacterium, had some benefit for
MDD, but we found no associations for these micro-
biota, possibly due to the synergistic effect of gut micro-
biome so that the influence of a particular taxon may be
different from multiple taxa [43]. Furthermore, these
clinical trials might draw biased conclusions because of
small sample sizes (ranging from 17 to 110) or short-
term effects (ranging from 3 to 24 weeks). Therefore, a
large and long-term RCT in a well-characterized popula-
tion using probiotic capsules containing specific micro-
biota might provide further evidence for the gut-brain
axis in these disorders. Importantly, epidemiological
study indicated that elevated Enterobacteriales was also
associated with a higher risk of ASD, suggesting that the
same changes in intestinal microbiota composition
might lead to different outcomes due to gene-gene inter-
actions and gene-environment interactions [44]. Al-
though our results showed no significant association for
Gammaproteobacteria and MDD, animal models found
increased levels of Gammaproteobacteria were also asso-
ciated with higher MDD risk and fluoxetine treatment
was effective, implying strong correlations between gut
microbiota and anxiety- and depression-like behaviors
[45].
The serotonin hypothesis of SCZ originated from earl-
ier studies of interactions between the hallucinogenic
drug D-lysergic acid diethylamide and serotonin in per-
ipheral systems. However, direct evidence of serotoner-
gic dysfunction in the pathogenesis of SCZ remains
unclear [46]. According to the principle of brain plasti -
city, glutamate signals are destroyed by serotonergic
overdrive, leading to neuronal hypometabolism, synaptic
atrophy, and gray matter loss in the end [47]. Our find-
ings that genetically increased serotonin levels was po-
tentially related to a high risk of SCZ using a MR
Fig. 3 Causal effect of serotonin with the risk of SCZ. a
Schematic representation of the MR analysis results: genetically
determined higher
serotonin plasma levels were potentially associated with a
higher risk of SCZ. b The odds ratios (95% confidence interval)
for SCZ per 10 units
increase in serotonin, as estimated in the inverse-variance
weighted, weighted mode, weighted median, and MR-Egger MR
analysis. The intercept
of MR-Egger can be interpreted as a test of overall unbalanced
horizontal pleiotropy. c The scatter plot represents instruments
association
including SCZ associations (y-axis) against instrument
serotonin associations (x-axis). The tunnel plot represents
instrument precision (i.e.,
instrument SCZ regression coefficients divided by the
correspondent instrument serotonin SEs) (y-axis) against
individual instrument ratio
estimates in log odds ratio of SCZ (x-axis). βIV indicates odds
ratio estimate per 1-ln 10 units increment in serotonin levels.
SCZ, schizophrenia
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 6 of 9
approach supported such hypothesis. Importantly, En-
terobacteriaceae family and Enterobacteriales order can
produce SCFAs (e.g., acetic acid and formic acid) in
carbohydrate fermentation, thus inducing serotonin bio-
synthesis by enterochromaffin cells which are the major
producers of serotonin, and ultimately increasing the
risk of SCZ [48, 49]. Our novel findings highlighted the
potentially important role of gut microbiota-related neu-
rotransmitters in effective and benign therapies of psy-
chiatric disorders.
Furthermore, we also found that neuropsychiatric disor-
ders might alter the composition of gut microbiota. Our
findings were consistent with a small case-control study (n
= 50) suggesting that Erysipelotrichaceae family were all
less abundant in patients with AD [5]. An observational
study showed that Porphyromonadaceae were associated
with poor cognitive performance, partly consistent with our
results [50]. However, the results from animal studies are
conflicting. Although several animal studies suggested that
anti-AD microbes, such as Erysipelotrichiaceae, decreased
in mouse models with AD, and Porphyromonadaceae in-
creased in aged mice [28, 51], other animal studies showed
that the relative abundance of Erysipelotrichiaceae was
positively correlated with AD [52, 53]. Therefore, the asso-
ciation of neuropsychiatric disorders with specific gut
microbiota requires further study. It is universally accepted
that the CNS modulates gut microbiota compositions
mainly through hypothalamic-pituitary-adrenal (HPA) axis,
or classical neurotransmitters liberated by neuronal efferent
activation, which explains the microbiota-host crosstalk in
neuropsychiatric disorders from another direction [54].
Additionally, it is plausible that alterations in gut
microbiota and related metabolites would lead to a sys-
temic change in inflammation that may contribute to
the neuroinflammation in AD, MDD, and SCZ. Increas-
ing evidence suggests that bacteria populating the gut
microbiome may excrete large quantities of lipopolysac-
charides and amyloids, resulting in the pathogenesis of
AD during aging when the permeability of gastrointes-
tinal tract epithelium or blood-brain barrier increases
[55]. Recent research has indicated that gut inflamma-
tion can induce activation of microglia and the kynure-
nine pathway, which activate systemic inflammation-
inducing depressive or schizophrenic symptoms [56, 57].
Therefore, more studies are required to explore the
mechanisms underlying the relationships of inflamma-
tion with the gut microbiota-brain axis and its relations
with AD, MDD and SCZ.
Strengths of the present study included the bi-directional
MR design and the use of summary-level data from thus far
the largest GWASs. This design generally avoided bias due
to reverse causation and confounding to obtain accurate
results under MR assumptions. In addition, consistent re-
sults from several sensitivity analyses including the use of
weighted mode, weighted median, and MR-egger methods
indicate robustness of our findings. However, several limita-
tions merit consideration. First, our results did not survive
a strict Bonferroni correction adjusting for multiple com-
parisons, whereas as a hypothesis-driven approach, the MR
study with some biological evidence was used to test epide-
miologically established associations, regardless of Bonfer -
roni corrected P values. Second, we used limited number of
gut microbiota SNPs as instrumental variables; we cannot
exclude that our findings might have been affected by weak
instrument bias, although all genetic instruments were as-
sociated with the exposure (F-statistic > 10). Third, statis-
tical power was limited for associations of neuropsychiatric
disorders with gut microbiota, so we cannot exclude type II
error as an explanation for the null results completely. Lar -
ger GWASs of gut microbiota are required to provide suffi -
cient statistical power. However, the power was strong
enough for the effect of gut microbiota on these disorders,
which was our main findings in the present study. Fourth,
our results were restricted to European ancestry. Replica-
tion with functionally relevant genetic prediction of gut
microbiota is warranted given the substantial difference in
gut microbiota composition among different populations.
Fifth, the 16S rRNA gene sequencing only permit reso-
lution from the genus to the phylum level rather than at a
more specific level, resulting in biased results if some spe-
cific species contributed to neuropsychiatric disorders. Fi -
nally, gut microbiota might be influenced by environmental
factors such as dietary habits or health status, which led to
lower variance explained by genetic instruments. However,
we could not test whether genetic instruments are associ-
ated with these confounders such as diet or lifestyle infor -
mation in the present study where such information is not
available.
Conclusions
In summary, our findings supported several potential as-
sociations between specific gut microbiota and neuro-
psychiatric disorders and highlighted the important roles
of microbial neurotransmitters such as GABA and sero-
tonin in microbiota-host crosstalk in neuropsychiatric
disorders. Further investigations in understanding the
underlying mechanisms of gut microbiota in the devel-
opment of neuropsychiatric disorders are required.
Supplementary information
Supplementary information accompanies this paper at
https://doi.org/10.
1186/s12974-020-01961-8.
Additional file 1: Figure S1. Odds ratio for association of
genetically
predicted gut microbiota with neuropsychological diseases.
Table S1.
Characteristics of selected SNPs for core gut microbiota. Table
S2.
Characteristics of selected SNPs for gut metabolites. Table S3.
Characteristics of selected SNPs for neuropsychological
diseases. Table
S4. Description of the diagnostic assessment for
neuropsychological
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 7 of 9
https://doi.org/10.1186/s12974-020-01961-8
https://doi.org/10.1186/s12974-020-01961-8
diseases. Table S5. Associations between genetically predicted
gut
microbiota and neuropsychological diseases in sensitivity
analyses. Table
S6. Associations between genetically predicted gut microbiota
and
neuropsychological diseases in a leave-one-out approach. Table
S7. As-
sociations between genetically predicted metabolites and
neuropsycho-
logical diseases using IVW method. Table S8. MR Power
calculation for
detecting significant (P < 7.6 × 10-4) causal effect (OR = 1.2)
of gut micro-
biome on the risk of AD, MDD, and SCZ. Table S9. Effect
estimates for
association of genetically predicted neuropsychological diseases
with gut
microbiota using four Mendelian randomization methods.
Abbreviations
AD: Alzheimer’s disease; MDD: Major depressive disorder;
SCZ: Schizophrenia;
MR: Mendelian randomization; GWAS: Genome-wide
association study;
GABA: γ-Aminobutyric acid; SCFA: Short-chain fatty acid;
BHB: β-
Hydroxybutyric acid; TMAO: Trimethylamine N-oxide; SNP:
Single nucleotide
polymorphism; IGAP: International Genomics of Alzheimer’s
Project;
PGC29: Psychiatric Genomics Consortium 29; LD: Linkage
disequilibrium;
IVW: Inverse variance weighting; CNS: Central nervous
system;
HPA: Hypothalamic-pituitary-adrenal; ASD: Autistic spectrum
disorder
Acknowledgements
The PopGen 2.0 network (P2N) is supported by a grant from the
German
Federal Ministry for Education and Research (01EY1103). We
thank Drs. Andre
Franke and Wolfgang Lieb for sharing the GWAS summary data
for beta
diversity and bacterial abundance from published paper (Nat
Genet. 2016
Nov; 48(11): 1396-1406.)
Authors’ contributions
ZZ, LQ, and TH designed the research. ZZ and TH had full
access to all the
data in the study and take responsibility for the integrity of the
data and the
accuracy of the data analysis. ZZ, LQ, and TH wrote the paper
and performed
the data analysis. All authors contributed to the statistical
analysis, critically
reviewed the manuscript during the writing process, and
approved the final
version to be published. ZZ and TH are the guarantors for the
study.
Funding
The study was supported by grants from the National Key
Research and
Development Project (2019YFC2003400), the Peking University
Start-up Grant
(BMU2018YJ002), High-performance Computing Platform of
Peking Univer-
sity, and the China-Canada Key Lab of Nutrition and Health at
Beijing Tech-
nology and Business University- Grant: 88442Y0033. The
funding organization
had no role in the preparation of the manuscript.
Availability of data and materials
All data used in the present study were obtained from genome-
wide associ-
ation study summary statistics which were publicly released by
genetic
consortia.
Ethics approval and consent to participate
Contributing studies received ethical approval from their
respective
institutional review boards.
Consent for publication
Not applicable.
Competing interests
All authors declare no support from companies for the
submitted work; no
relationships with companies that might have an interest in the
submitted
work in the previous 3 years; no spouses, partners, or children
that have
financial relationships that may be relevant to the submitted
work; and no
non-financial interests that may be relevant to the submitted
work.
Author details
1Department of Epidemiology & Biostatistics, School of Public
Health, Peking
University, 38 Xueyuan Road, Beijing 100191, China.
2Department of
Epidemiology, School of Public Health and Tropical Medicine,
Tulane
University, New Orleans, LA, USA. 3Department of Nutrition,
Harvard T.H.
Chan School of Public Health, Boston, MA, USA. 4Department
of Global
Health, School of Public Health, Peking University, Beijing
100191, China. 5Key
Laboratory of Molecular Cardiovascular Sciences (Peking
University), Ministry
of Education, Beijing 100191, China. 6Center for Intelligent
Public Health,
Institute for Artificial Intelligence, Peking University, Beijing
100191, China.
Received: 2 July 2020 Accepted: 23 September 2020
References
1. Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang
J, Tito RY,
Schiweck C, Kurilshikov A, Joossens M, Wijmenga C, et al.
The neuroactive
potential of the human gut microbiota in quality of life and
depression. Nat
Microbiol. 2019;4(4):623–32.
2. Fung TC, Olson CA, Hsiao EY. Interactions between the
microbiota, immune
and nervous systems in health and disease. Nat Neurosci.
2017;20(2):145–55.
3. Caspani G, Swann J. Small talk: microbial metabolites
involved in the
signaling from microbiota to brain. Curr Opin Pharmacol.
2019;48:99–106.
4. van de Wouw M, Boehme M, Lyte JM, Wiley N, Strain C,
O'Sullivan O, Clarke
G, Stanton C, Dinan TG, Cryan JF. Short-chain fatty acids:
microbial
metabolites that alleviate stress-induced brain-gut axis
alterations. J Physiol.
2018;596(20):4923–44.
5. Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ,
Merluzzi AP, Johnson SC,
Carlsson CM, Asthana S, Zetterberg H, Blennow K, et al. Gut
microbiome
alterations in Alzheimer’s disease. Sci Rep. 2017;7(1):13537.
6. Shen Y, Xu J, Li Z, Huang Y, Yuan Y, Wang J, Zhang M, Hu
S, Liang Y.
Analysis of gut microbiota diversity and auxiliary diagnosis as a
biomarker in
patients with schizophrenia: a cross-sectional study. Schizophr
Res. 2018;197:
470–7.
7. Naseribafrouei A, Hestad K, Avershina E, Sekelja M,
Linlokken A, Wilson R,
Rudi K. Correlation between the human fecal microbiota and
depression.
Neurogastroenterol Motil. 2014;26(8):1155–62.
8. Strandwitz P, Kim KH, Terekhova D, Liu JK, Sharma A,
Levering J, McDonald
D, Dietrich D, Ramadhar TR, Lekbua A, et al. GABA-
modulating bacteria of
the human gut microbiota. Nat Microbiol. 2019;4(3):396–403.
9. Strandwitz P. Neurotransmitter modulation by the gut
microbiota. Brain Res.
2018;1693(Pt B):128–33.
10. Huang D, Liu D, Yin J, Qian T, Shrestha S, Ni H.
Glutamate-glutamine and
GABA in brain of normal aged and patients with cognitive
impairment. Eur
Radiol. 2017;27(7):2698–705.
11. Peitl V, Štefanović M, Karlović D. Depressive symptoms in
schizophrenia and
dopamine and serotonin gene polymorphisms. Prog
Neuropsychopharmacol Biol Psychiatry. 2017;77:209–15.
12. Manyevitch R, Protas M, Scarpiello S, Deliso M, Bass B,
Nanajian A, Chang M,
Thompson SM, Khoury N, Gonnella R, et al. Evaluation of
metabolic and
synaptic dysfunction hypotheses of Alzheimer’s disease (AD): a
meta-
analysis of CSF markers. Curr Alzheimer Res. 2018;15(2):164–
81.
13. Ertugrul A, Ucar G, Basar K, Demir B, Yabanoglu S, Ulug
B. Influence of
clozapine on platelet serotonin, monoamine oxidase and plasma
serotonin
levels. Psychiatry Res. 2007;149(1-3):49–57.
14. Peitl V, Vidrih B, Karlović Z, Getaldić B, Peitl M, Karlović
D. Platelet serotonin
concentration and depressive symptoms in patients with
schizophrenia.
Psychiatry Res. 2016;239:105–10.
15. Emdin CA, Khera AV, Kathiresan S. Mendelian
randomization. JAMA. 2017;
318(19):1925–6.
16. Sanna S, van Zuydam NR, Mahajan A, Kurilshikov A, Vich
Vila A, Võsa U,
Mujagic Z, Masclee AAM, Jonkers DMAE, Oosting M, et al.
Causal
relationships among the gut microbiome, short-chain fatty acids
and
metabolic diseases. Nat Genet. 2019;51(4):600–5.
17. Turpin W, Espin-Garcia O, Xu W, Silverberg MS, Kevans
D, Smith MI,
Guttman DS, Griffiths A, Panaccione R, Otley A, et al.
Association of host
genome with intestinal microbial composition in a large healthy
cohort. Nat
Genet. 2016;48(11):1413–7.
18. Goodrich JK, Davenport ER, Beaumont M, Jackson MA,
Knight R, Ober C,
Spector TD, Bell JT, Clark AG, Ley RE. Genetic determinants
of the gut
microbiome in UK twins. Cell Host Microbe. 2016;19(5):731–
43.
19. Wang J, Thingholm LB, Skiecevičienė J, Rausch P,
Kummen M, Hov JR,
Degenhardt F, Heinsen F-A, Rühlemann MC, Szymczak S, et al.
Genome-
wide association analysis identifies variation in vitamin D
receptor and other
host factors influencing the gut microbiota. Nat Genet.
2016;48(11):1396–
406.
20. Rhee EP, Ho JE, Chen MH, Shen D, Cheng S, Larson MG,
Ghorbani A, Shi X,
Helenius IT, O'Donnell CJ, et al. A genome-wide association
study of the
Zhuang et al. Journal of Neuroinfla mmation (2020)
17:288 Page 8 of 9
human metabolome in a community-based cohort. Cell Metab.
2013;18(1):
130–43.
21. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims
R, Bellenguez C,
DeStafano AL, Bis JC, Beecham GW, Grenier-Boley B, et al.
Meta-analysis of
74,046 individuals identifies 11 new susceptibility loci for
Alzheimer’s
disease. Nat Genet. 2013;45(12):1452–8.
22. Wray NR, Ripke S, Mattheisen M, Trzaskowski M, Byrne
EM, Abdellaoui A,
Adams MJ, Agerbo E, Air TM, Andlauer TMF, et al. Genome-
wide association
analyses identify 44 risk variants and refine the genetic
architecture of major
depression. Nat Genet. 2018;50(5):668–81.
23. Ripke S, O'Dushlaine C, Chambert K, Moran JL, Kähler AK,
Akterin S, Bergen
SE, Collins AL, Crowley JJ, Fromer M, et al. Genome-wide
association
analysis identifies 13 new risk loci for schizophrenia. Nat
Genet. 2013;45(10):
1150–9.
24. Sabokdast M, Habibi-Rezaei M, Moosavi-Movahedi AA,
Ferdousi M,
Azimzadeh-Irani E, Poursasan N. Protection by beta-
Hydroxybutyric acid
against insulin glycation, lipid peroxidation and microglial cell
apoptosis.
Daru. 2015;23:42.
25. Zalar B, Haslberger A, Peterlin B. The role of microbiota in
depression - a
brief review. Psychiatr Danub. 2018;30(2):136–41.
26. Bowden J, Davey Smith G, Haycock PC, Burgess S.
Consistent estimation in
Mendelian randomization with some invalid instruments using a
weighted
median estimator. Genet Epidemiol. 2016;40(4):304–14.
27. Hartwig FP, Davey Smith G, Bowden J. Robust inference in
summary data
Mendelian randomization via the zero modal pleiotropy
assumption. Int J
Epidemiol. 2017;46(6):1985–98.
28. Xin Y, Diling C, Jian Y, Ting L, Guoyan H, Hualun L,
Xiaocui T, Guoxiao L, Ou
S, Chaoqun Z, et al. Effects of oligosaccharides from Morinda
officinalis on
gut microbiota and metabolome of APP/PS1 transgenic mice.
Front Neurol.
2018;9:412.
29. Park J-Y, Choi J, Lee Y, Lee J-E, Lee E-H, Kwon H-J, Yang
J, Jeong B-R, Kim Y-
K, Han P-L. Metagenome analysis of bodily microbiota in a
mouse model of
Alzheimer disease using bacteria-derived membrane vesicles in
blood. Exp
Neurobiol. 2017;26(6):369–79.
30. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci
V, McCormick BA.
Alzheimer’s disease microbiome is associated with
dysregulation of the anti-
inflammatory P-glycoprotein pathway. mBio.
2019;10(3):e00632–19.
31. Li B, He Y, Ma J, Huang P, Du J, Cao L, Wang Y, Xiao Q,
Tang H, Chen S.
Mild cognitive impairment has similar alterations as
Alzheimer’s disease in
gut microbiota. Alzheimers Dement. 2019;15(10):1357–66.
32. Gueli MC, Taibi G. Alzheimer’s disease: amino acid levels
and brain
metabolic status. Neurol Sci. 2013;34(9):1575–9.
33. Jiménez-Jiménez FJ, Molina JA, Gómez P, Vargas C, de
Bustos F, Benito-
León J, Tallón-Barranco A, Ortí-Pareja M, Gasalla T, Arenas J.
Neurotransmitter amino acids in cerebrospinal fluid of patients
with
Alzheimer’s disease. J Neural Transm (Vienna). 1998;105(2-
3):269–77.
34. Liu F, Li J, Wu F, Zheng H, Peng Q, Zhou H. Altered
composition and
function of intestinal microbiota in autism spectrum disorders: a
systematic
review. Transl Psychiatry. 2019;9(1):43.
35. de JR De-Paula V, Forlenza AS, Forlenza OV. Relevance of
gutmicrobiota in
cognition, behaviour and Alzheimer’s disease. Pharmacol Res.
2018;136:29–34.
36. Rissman RA, De Blas AL, Armstrong DM. GABA(A)
receptors in aging and
Alzheimer’s disease. J Neurochem. 2007;103(4):1285–92.
37. Govindpani K, Calvo-Flores Guzmán B, Vinnakota C,
Waldvogel HJ, Faull RL,
Kwakowsky A. Towards a better understanding of GABAergic
remodeling in
Alzheimer’s disease. Int J Mol Sci. 2017;18(8):1813.
38. Liu X, Zheng H, Lu R, Huang H, Zhu H, Yin C, Mo Y, Wu
J, Liu X, Deng M,
et al. Intervening effects of total alkaloids of Corydalis saxicola
Bunting on
rats with antibiotic-induced gut microbiota dysbiosis based on
16S rRNA
gene sequencing and untargeted metabolomics analyses. Front
Microbiol.
2019;10:1151.
39. Bergin DH, Jing Y, Zhang H, Liu P. A single
intracerebroventricular Aβ25-35
infusion leads to prolonged alterations in arginine metabolism
in the rat
hippocampus and prefrontal cortex. Neuroscience.
2015;298:367–79.
40. Nguyen TT, Kosciolek T, Maldonado Y, Daly RE, Martin
AS, McDonald D,
Knight R, Jeste DV. Differences in gut microbiome composition
between
persons with chronic schizophrenia and healthy comparison
subjects.
Schizophr Res. 2019;204:23–9.
41. Cao G, Tao F, Hu Y, Li Z, Zhang Y, Deng B, Zhan X.
Positive effects of a
Clostridium butyricum-based compound probiotic on growth
performance,
immune responses, intestinal morphology, hypothalamic
neurotransmitters,
and colonic microbiota in weaned piglets. Food Funct.
2019;10(5):2926–34.
42. Opmeer EM, Kortekaas R, Aleman A. Depression and the
role of genes
involved in dopamine metabolism and signalling. Prog
Neurobiol. 2010;
92(2):112–33.
43. Barbosa RSD, Vieira-Coelho MA. Probiotics and prebiotics:
focus on
psychiatric disorders - a systematic review. Nutr Rev.
2020;78(6):437-50.
44. Liu S, Li E, Sun Z, Fu D, Duan G, Jiang M, Yu Y, Mei L,
Yang P, Tang Y, et al.
Altered gut microbiota and short chain fatty acids in Chinese
children with
autism spectrum disorder. Sci Rep. 2019;9(1):287.
45. Sun L, Zhang H, Cao Y, Wang C, Zhao C, Wang H, Cui G,
Wang M, Pan Y, Shi Y,
et al. Fluoxetine ameliorates dysbiosis in a depression model
induced by
chronic unpredicted mild stress in mice. Int J Med Sci.
2019;16(9):1260–70.
46. Yang AC, Tsai S-J. New targets for schizophrenia treatment
beyond the
dopamine hypothesis. Int J Mol Sci. 2017;18(8):1689.
47. Abi-Dargham A, Laruelle M, Aghajanian GK, Charney D,
Krystal J. The role of
serotonin in the pathophysiology and treatment of
schizophrenia. J
Neuropsychiatry Clin Neurosci. 1997;9(1):1–17.
48. Ge X, Pan J, Liu Y, Wang H, Zhou W, Wang X. Intestinal
crosstalk between
microbiota and serotonin and its impact on gut motility. Curr
Pharm
Biotechnol. 2018;19(3):190–5.
49. Blander JM, Longman RS, Iliev ID, Sonnenberg GF, Artis
D. Regulation of
inflammation by microbiota interactions with the host. Nat
Immunol. 2017;
18(8):851–60.
50. Bajaj JS, Ahluwalia V, Steinberg JL, Hobgood S, Boling
PA, Godschalk M,
Habib S, White MB, Fagan A, Gavis EA, et al. Elderly patients
have an altered
gut-brain axis regardless of the presence of cirrhosis. Sci Rep.
2016;6:38481.
51. Du C-T, Gao W, Ma K, Yu S-X, Li N, Yan S-Q, Zhou F-H,
Liu Z-Z, Chen W, Lei
L-C, et al. MicroRNA-146a deficiency protects against Listeria
monocytogenes infection by modulating the gut microbiota. Int
J Mol Sci.
2018;19(4):993.
52. Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc
Coy KD, Frisoni G,
Neher JJ, Fak F, Jucker M, Lasser T, et al. Reduction of Abeta
amyloid
pathology in APPPS1 transgenic mice in the absence of gut
microbiota. Sci
Rep. 2017;7:41802.
53. Bauerl C, Collado MC, Diaz Cuevas A, Vina J, Perez
Martinez G. Shifts in gut
microbiota composition in an APP/PSS1 transgenic mouse
model of
Alzheimer’s disease during lifespan. Lett Appl Microbiol.
2018;66(6):464–71.
54. Montiel-Castro AJ, González-Cervantes RM, Bravo-Ruiseco
G, Pacheco-López
G. The microbiota-gut-brain axis: neurobehavioral correlates,
health and
sociality. Front Integr Neurosci. 2013;7:70.
55. Lin L, Zheng LJ, Zhang LJ. Neuroinflammation, gut
microbiome, and
Alzheimer’s disease. Mol Neurobiol. 2018;55(11):8243–50.
56. Simkin DR. Microbiome and mental health, specifically as it
relates to
adolescents. Curr Psychiatry Rep. 2019;21(9):93.
57. Inta D, Lang UE, Borgwardt S, Meyer-Lindenberg A, Gass
P. Microglia
activation and schizophrenia: lessons from the effects of
minocycline on
postnatal neurogenesis, neuronal survival and synaptic pruning.
Schizophr
Bull. 2017;43(3):493–6.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional
claims in
published maps and institutional affiliations.
Zhuang et al. Journal of Neuroinflammation (2020)
17:288 Page 9 of 9
AbstractBackgroundMethodsResultsConclusionsBackgroundMet
hodsStudy design overviewData sources and instrumentsGut
microbiotaGut microbial metabolitesNeuropsychiatric
disordersStatistical analysisResultsAssociations of gut
microbiota and metabolites with neuropsychiatric
disordersAssociations of neuropsychiatric disorders with gut
microbiotaDiscussionConclusionsSupplementary
informationAbbreviationsAcknowledgementsAuthors’
contributionsFundingAvailability of data and materialsEthics
approval and consent to participateConsent for
publicationCompeting interestsAuthor
detailsReferencesPublisher’s Note
OPEN
ORIGINAL ARTICLE
Inflammasome signaling affects anxiety- and depressive-like
behavior and gut microbiome composition
M-L Wong1,2,9,10, A Inserra1,2,9, MD Lewis1,2, CA
Mastronardi3, L Leong4,5, J Choo4,5, S Kentish6, P Xie7,10, M
Morrison8, SL Wesselingh4,5,
GB Rogers4,5,10 and J Licinio1,2,10
The inflammasome is hypothesized to be a key mediator of the
response to physiological and psychological stressors, and its
dysregulation may be implicated in major depressive disorder.
Inflammasome activation causes the maturation of caspase-1
and
activation of interleukin (IL)-1β and IL-18, two
proinflammatory cytokines involved in
neuroimmunomodulation, neuroinflammation
and neurodegeneration. In this study, C57BL/6 mice with
genetic deficiency or pharmacological inhibition of caspase-1
were
screened for anxiety- and depressive-like behaviors, and
locomotion at baseline and after chronic stress. We found that
genetic
deficiency of caspase-1 decreased depressive- and anxiety-like
behaviors, and conversely increased locomotor activity and
skills.
Caspase-1 deficiency also prevented the exacerbation of
depressive-like behaviors following chronic stress. Furthermore,
pharmacological caspase-1 antagonism with minocycline
ameliorated stress-induced depressive-like behavior in wild-
type mice.
Interestingly, chronic stress or pharmacological inhibition of
caspase-1 per se altered the fecal microbiome in a very similar
manner.
When stressed mice were treated with minocycline, the
observed gut microbiota changes included increase in relative
abundance
of Akkermansia spp. and Blautia spp., which are compatible
with beneficial effects of attenuated inflammation and rebalance
of gut
microbiota, respectively, and the increment in Lachnospiracea
abundance was consistent with microbiota changes of caspase-1
deficiency. Our results suggest that the protective effect of
caspase-1 inhibition involves the modulation of the relationship
between stress and gut microbiota composition, and establishes
the basis for a gut microbiota–inflammasome–brain axis,
whereby
the gut microbiota via inflammasome signaling modulate
pathways that will alter brain function, and affect depressive-
and
anxiety-like behaviors. Our data also suggest that further
elucidation of the gut microbiota–inflammasome–brain axis may
offer
novel therapeutic targets for psychiatric disorders.
Molecular Psychiatry (2016) 21, 797–805;
doi:10.1038/mp.2016.46; published online 19 April 2016
INTRODUCTION
Increasing evidence suggests an involvement of neuroinflamma-
tory pathways in the etiopathophysiology of major depressive
disorder (MDD) and antidepressant response.1,2 Depressive
symptoms are underlined by increased levels of
proinflammatory
cytokines (that is, interleukin (IL)-1β and IL-6), decreased
levels of
anti-inflammatory cytokines (that is, IL-4 and IL-10) and are
associated with polymorphisms in inflammation-related
genes.3–5
IL-1 receptor type-I and its ligands are expressed in brain areas
relevant to stress response,6–8 and IL-1β signaling is
fundamental
in mediating the deleterious neurobehavioral and
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet
Examples of Nutrition ClaimsClaims about a popular diet

More Related Content

Similar to Examples of Nutrition ClaimsClaims about a popular diet

CHI-Obesity-White-Paper
CHI-Obesity-White-PaperCHI-Obesity-White-Paper
CHI-Obesity-White-PaperWill Zasadny
 
Essay For Health
Essay For HealthEssay For Health
Media Healthy Eating
Media Healthy EatingMedia Healthy Eating
Media Healthy Eatingionewright
 
COPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMS
COPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMSCOPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMS
COPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMSHarsha Aswani
 
Part 2 Baer Career Presentation June 2011
Part 2 Baer Career Presentation June 2011Part 2 Baer Career Presentation June 2011
Part 2 Baer Career Presentation June 2011Xcaret2010
 
Obesity case study.htm
Obesity case study.htmObesity case study.htm
Obesity case study.htmlindy paul
 
Probiotic symposium chennai 3 dec 2016
Probiotic symposium chennai 3 dec 2016Probiotic symposium chennai 3 dec 2016
Probiotic symposium chennai 3 dec 2016
neerjayakult
 
From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...
From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...
From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...
Norwich Research Park
 
A METHODOLOGICAL ANALYSIS ON OBESITY
A METHODOLOGICAL ANALYSIS ON OBESITYA METHODOLOGICAL ANALYSIS ON OBESITY
A METHODOLOGICAL ANALYSIS ON OBESITY
SSR Institute of International Journal of Life Sciences
 
Nutrition Vs Stress
Nutrition Vs StressNutrition Vs Stress
Nutrition Vs StressBJuszczyk
 
Nutrition Vs Stress
Nutrition Vs StressNutrition Vs Stress
Nutrition Vs StressPJuszczyk
 
PPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.pptPPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.ppt
InnaMutmainnahMusa
 
Running header THE MENACE OF OBESTIY1The Me.docx
Running header THE MENACE OF OBESTIY1The Me.docxRunning header THE MENACE OF OBESTIY1The Me.docx
Running header THE MENACE OF OBESTIY1The Me.docx
anhlodge
 
Children's Health
Children's HealthChildren's Health

Similar to Examples of Nutrition ClaimsClaims about a popular diet (14)

CHI-Obesity-White-Paper
CHI-Obesity-White-PaperCHI-Obesity-White-Paper
CHI-Obesity-White-Paper
 
Essay For Health
Essay For HealthEssay For Health
Essay For Health
 
Media Healthy Eating
Media Healthy EatingMedia Healthy Eating
Media Healthy Eating
 
COPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMS
COPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMSCOPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMS
COPING STRATEGIES FOR PHYSICAL HEALTH PROBLEMS
 
Part 2 Baer Career Presentation June 2011
Part 2 Baer Career Presentation June 2011Part 2 Baer Career Presentation June 2011
Part 2 Baer Career Presentation June 2011
 
Obesity case study.htm
Obesity case study.htmObesity case study.htm
Obesity case study.htm
 
Probiotic symposium chennai 3 dec 2016
Probiotic symposium chennai 3 dec 2016Probiotic symposium chennai 3 dec 2016
Probiotic symposium chennai 3 dec 2016
 
From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...
From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...
From  Nutrigenomics to Systems Nutrition - The role of nutrition in metabolic...
 
A METHODOLOGICAL ANALYSIS ON OBESITY
A METHODOLOGICAL ANALYSIS ON OBESITYA METHODOLOGICAL ANALYSIS ON OBESITY
A METHODOLOGICAL ANALYSIS ON OBESITY
 
Nutrition Vs Stress
Nutrition Vs StressNutrition Vs Stress
Nutrition Vs Stress
 
Nutrition Vs Stress
Nutrition Vs StressNutrition Vs Stress
Nutrition Vs Stress
 
PPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.pptPPT nutrigenomics obesity in nutrition.ppt
PPT nutrigenomics obesity in nutrition.ppt
 
Running header THE MENACE OF OBESTIY1The Me.docx
Running header THE MENACE OF OBESTIY1The Me.docxRunning header THE MENACE OF OBESTIY1The Me.docx
Running header THE MENACE OF OBESTIY1The Me.docx
 
Children's Health
Children's HealthChildren's Health
Children's Health
 

More from BetseyCalderon89

MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docxMANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
BetseyCalderon89
 
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docxManage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
BetseyCalderon89
 
Make sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docxMake sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docx
BetseyCalderon89
 
Make sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docxMake sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docx
BetseyCalderon89
 
make sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docxmake sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docx
BetseyCalderon89
 
make two paragraphs on diffences and similiarties religous belifs .docx
make two paragraphs on diffences and similiarties  religous belifs .docxmake two paragraphs on diffences and similiarties  religous belifs .docx
make two paragraphs on diffences and similiarties religous belifs .docx
BetseyCalderon89
 
Make a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docxMake a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docx
BetseyCalderon89
 
Make a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docxMake a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docx
BetseyCalderon89
 
Make a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docxMake a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docx
BetseyCalderon89
 
Make sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docxMake sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docx
BetseyCalderon89
 
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docxMajor Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
BetseyCalderon89
 
Major earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docxMajor earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docx
BetseyCalderon89
 
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docxMajor Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
BetseyCalderon89
 
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docxMaintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
BetseyCalderon89
 
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docxMaintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
BetseyCalderon89
 
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docxMacro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
BetseyCalderon89
 
Macromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docxMacromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docx
BetseyCalderon89
 
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docxM7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
BetseyCalderon89
 
Madison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docxMadison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docx
BetseyCalderon89
 
Main content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docxMain content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docx
BetseyCalderon89
 

More from BetseyCalderon89 (20)

MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docxMANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
MANAGEGIAL ECONOMICS AND ORGANIZATIONAL ARCHITECTURE 5Th Edition .docx
 
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docxManage Resourcesfor Practicum Change ProjectYou are now half-w.docx
Manage Resourcesfor Practicum Change ProjectYou are now half-w.docx
 
Make sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docxMake sure you put it in your own words and references for each pleas.docx
Make sure you put it in your own words and references for each pleas.docx
 
Make sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docxMake sure you take your time and provide complete answers. Two or th.docx
Make sure you take your time and provide complete answers. Two or th.docx
 
make sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docxmake sure is 100 original not copythis first questionDiscuss .docx
make sure is 100 original not copythis first questionDiscuss .docx
 
make two paragraphs on diffences and similiarties religous belifs .docx
make two paragraphs on diffences and similiarties  religous belifs .docxmake two paragraphs on diffences and similiarties  religous belifs .docx
make two paragraphs on diffences and similiarties religous belifs .docx
 
Make a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docxMake a list of your own personality traits and then address the foll.docx
Make a list of your own personality traits and then address the foll.docx
 
Make a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docxMake a list of your own personality traits and then address the .docx
Make a list of your own personality traits and then address the .docx
 
Make a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docxMake a list of people you consider to be your close friend. For each.docx
Make a list of people you consider to be your close friend. For each.docx
 
Make sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docxMake sure questions and references are included! Determine how s.docx
Make sure questions and references are included! Determine how s.docx
 
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docxMajor Paper #2--The Personal Narrative EssayA narrative is simpl.docx
Major Paper #2--The Personal Narrative EssayA narrative is simpl.docx
 
Major earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docxMajor earthquakes and volcano eruptions occurred long before there w.docx
Major earthquakes and volcano eruptions occurred long before there w.docx
 
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docxMajor Paper #1-The Point of View Essay Deadline October 29, 2.docx
Major Paper #1-The Point of View Essay Deadline October 29, 2.docx
 
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docxMaintenance and TroubleshootingDescribe the maintenance procedures.docx
Maintenance and TroubleshootingDescribe the maintenance procedures.docx
 
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docxMaintaining the Loyalty of StakeholdersTo maintain political, gove.docx
Maintaining the Loyalty of StakeholdersTo maintain political, gove.docx
 
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docxMacro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
Macro Paper Assignment - The Eurozone Crisis - DueOct 22, 2015.docx
 
Macromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docxMacromolecules are constructed as a result of covalent forced; howev.docx
Macromolecules are constructed as a result of covalent forced; howev.docx
 
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docxM7A1 Resolving ConflictIf viewing this through the Assignment too.docx
M7A1 Resolving ConflictIf viewing this through the Assignment too.docx
 
Madison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docxMadison is interested in how many of the children in.docx
Madison is interested in how many of the children in.docx
 
Main content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docxMain content areaBased on the readings this week with special at.docx
Main content areaBased on the readings this week with special at.docx
 

Recently uploaded

A Strategic Approach: GenAI in Education
A Strategic Approach: GenAI in EducationA Strategic Approach: GenAI in Education
A Strategic Approach: GenAI in Education
Peter Windle
 
Thesis Statement for students diagnonsed withADHD.ppt
Thesis Statement for students diagnonsed withADHD.pptThesis Statement for students diagnonsed withADHD.ppt
Thesis Statement for students diagnonsed withADHD.ppt
EverAndrsGuerraGuerr
 
Language Across the Curriculm LAC B.Ed.
Language Across the  Curriculm LAC B.Ed.Language Across the  Curriculm LAC B.Ed.
Language Across the Curriculm LAC B.Ed.
Atul Kumar Singh
 
Model Attribute Check Company Auto Property
Model Attribute  Check Company Auto PropertyModel Attribute  Check Company Auto Property
Model Attribute Check Company Auto Property
Celine George
 
Acetabularia Information For Class 9 .docx
Acetabularia Information For Class 9  .docxAcetabularia Information For Class 9  .docx
Acetabularia Information For Class 9 .docx
vaibhavrinwa19
 
Embracing GenAI - A Strategic Imperative
Embracing GenAI - A Strategic ImperativeEmbracing GenAI - A Strategic Imperative
Embracing GenAI - A Strategic Imperative
Peter Windle
 
CLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCE
CLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCECLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCE
CLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCE
BhavyaRajput3
 
1.4 modern child centered education - mahatma gandhi-2.pptx
1.4 modern child centered education - mahatma gandhi-2.pptx1.4 modern child centered education - mahatma gandhi-2.pptx
1.4 modern child centered education - mahatma gandhi-2.pptx
JosvitaDsouza2
 
Unit 8 - Information and Communication Technology (Paper I).pdf
Unit 8 - Information and Communication Technology (Paper I).pdfUnit 8 - Information and Communication Technology (Paper I).pdf
Unit 8 - Information and Communication Technology (Paper I).pdf
Thiyagu K
 
The Challenger.pdf DNHS Official Publication
The Challenger.pdf DNHS Official PublicationThe Challenger.pdf DNHS Official Publication
The Challenger.pdf DNHS Official Publication
Delapenabediema
 
Home assignment II on Spectroscopy 2024 Answers.pdf
Home assignment II on Spectroscopy 2024 Answers.pdfHome assignment II on Spectroscopy 2024 Answers.pdf
Home assignment II on Spectroscopy 2024 Answers.pdf
Tamralipta Mahavidyalaya
 
Instructions for Submissions thorugh G- Classroom.pptx
Instructions for Submissions thorugh G- Classroom.pptxInstructions for Submissions thorugh G- Classroom.pptx
Instructions for Submissions thorugh G- Classroom.pptx
Jheel Barad
 
Honest Reviews of Tim Han LMA Course Program.pptx
Honest Reviews of Tim Han LMA Course Program.pptxHonest Reviews of Tim Han LMA Course Program.pptx
Honest Reviews of Tim Han LMA Course Program.pptx
timhan337
 
CACJapan - GROUP Presentation 1- Wk 4.pdf
CACJapan - GROUP Presentation 1- Wk 4.pdfCACJapan - GROUP Presentation 1- Wk 4.pdf
CACJapan - GROUP Presentation 1- Wk 4.pdf
camakaiclarkmusic
 
"Protectable subject matters, Protection in biotechnology, Protection of othe...
"Protectable subject matters, Protection in biotechnology, Protection of othe..."Protectable subject matters, Protection in biotechnology, Protection of othe...
"Protectable subject matters, Protection in biotechnology, Protection of othe...
SACHIN R KONDAGURI
 
Palestine last event orientationfvgnh .pptx
Palestine last event orientationfvgnh .pptxPalestine last event orientationfvgnh .pptx
Palestine last event orientationfvgnh .pptx
RaedMohamed3
 
The geography of Taylor Swift - some ideas
The geography of Taylor Swift - some ideasThe geography of Taylor Swift - some ideas
The geography of Taylor Swift - some ideas
GeoBlogs
 
Lapbook sobre os Regimes Totalitários.pdf
Lapbook sobre os Regimes Totalitários.pdfLapbook sobre os Regimes Totalitários.pdf
Lapbook sobre os Regimes Totalitários.pdf
Jean Carlos Nunes Paixão
 
TESDA TM1 REVIEWER FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...
TESDA TM1 REVIEWER  FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...TESDA TM1 REVIEWER  FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...
TESDA TM1 REVIEWER FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...
EugeneSaldivar
 
Biological Screening of Herbal Drugs in detailed.
Biological Screening of Herbal Drugs in detailed.Biological Screening of Herbal Drugs in detailed.
Biological Screening of Herbal Drugs in detailed.
Ashokrao Mane college of Pharmacy Peth-Vadgaon
 

Recently uploaded (20)

A Strategic Approach: GenAI in Education
A Strategic Approach: GenAI in EducationA Strategic Approach: GenAI in Education
A Strategic Approach: GenAI in Education
 
Thesis Statement for students diagnonsed withADHD.ppt
Thesis Statement for students diagnonsed withADHD.pptThesis Statement for students diagnonsed withADHD.ppt
Thesis Statement for students diagnonsed withADHD.ppt
 
Language Across the Curriculm LAC B.Ed.
Language Across the  Curriculm LAC B.Ed.Language Across the  Curriculm LAC B.Ed.
Language Across the Curriculm LAC B.Ed.
 
Model Attribute Check Company Auto Property
Model Attribute  Check Company Auto PropertyModel Attribute  Check Company Auto Property
Model Attribute Check Company Auto Property
 
Acetabularia Information For Class 9 .docx
Acetabularia Information For Class 9  .docxAcetabularia Information For Class 9  .docx
Acetabularia Information For Class 9 .docx
 
Embracing GenAI - A Strategic Imperative
Embracing GenAI - A Strategic ImperativeEmbracing GenAI - A Strategic Imperative
Embracing GenAI - A Strategic Imperative
 
CLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCE
CLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCECLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCE
CLASS 11 CBSE B.St Project AIDS TO TRADE - INSURANCE
 
1.4 modern child centered education - mahatma gandhi-2.pptx
1.4 modern child centered education - mahatma gandhi-2.pptx1.4 modern child centered education - mahatma gandhi-2.pptx
1.4 modern child centered education - mahatma gandhi-2.pptx
 
Unit 8 - Information and Communication Technology (Paper I).pdf
Unit 8 - Information and Communication Technology (Paper I).pdfUnit 8 - Information and Communication Technology (Paper I).pdf
Unit 8 - Information and Communication Technology (Paper I).pdf
 
The Challenger.pdf DNHS Official Publication
The Challenger.pdf DNHS Official PublicationThe Challenger.pdf DNHS Official Publication
The Challenger.pdf DNHS Official Publication
 
Home assignment II on Spectroscopy 2024 Answers.pdf
Home assignment II on Spectroscopy 2024 Answers.pdfHome assignment II on Spectroscopy 2024 Answers.pdf
Home assignment II on Spectroscopy 2024 Answers.pdf
 
Instructions for Submissions thorugh G- Classroom.pptx
Instructions for Submissions thorugh G- Classroom.pptxInstructions for Submissions thorugh G- Classroom.pptx
Instructions for Submissions thorugh G- Classroom.pptx
 
Honest Reviews of Tim Han LMA Course Program.pptx
Honest Reviews of Tim Han LMA Course Program.pptxHonest Reviews of Tim Han LMA Course Program.pptx
Honest Reviews of Tim Han LMA Course Program.pptx
 
CACJapan - GROUP Presentation 1- Wk 4.pdf
CACJapan - GROUP Presentation 1- Wk 4.pdfCACJapan - GROUP Presentation 1- Wk 4.pdf
CACJapan - GROUP Presentation 1- Wk 4.pdf
 
"Protectable subject matters, Protection in biotechnology, Protection of othe...
"Protectable subject matters, Protection in biotechnology, Protection of othe..."Protectable subject matters, Protection in biotechnology, Protection of othe...
"Protectable subject matters, Protection in biotechnology, Protection of othe...
 
Palestine last event orientationfvgnh .pptx
Palestine last event orientationfvgnh .pptxPalestine last event orientationfvgnh .pptx
Palestine last event orientationfvgnh .pptx
 
The geography of Taylor Swift - some ideas
The geography of Taylor Swift - some ideasThe geography of Taylor Swift - some ideas
The geography of Taylor Swift - some ideas
 
Lapbook sobre os Regimes Totalitários.pdf
Lapbook sobre os Regimes Totalitários.pdfLapbook sobre os Regimes Totalitários.pdf
Lapbook sobre os Regimes Totalitários.pdf
 
TESDA TM1 REVIEWER FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...
TESDA TM1 REVIEWER  FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...TESDA TM1 REVIEWER  FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...
TESDA TM1 REVIEWER FOR NATIONAL ASSESSMENT WRITTEN AND ORAL QUESTIONS WITH A...
 
Biological Screening of Herbal Drugs in detailed.
Biological Screening of Herbal Drugs in detailed.Biological Screening of Herbal Drugs in detailed.
Biological Screening of Herbal Drugs in detailed.
 

Examples of Nutrition ClaimsClaims about a popular diet

  • 1. Examples of Nutrition Claims Claims about a popular diet that is supposed to change your body, reverse a disease, or dramatically improve your health or performance in some way. Claims about a particular food, beverage or dietary supplement that is supposed to help you lose weight, gain muscle, boost immunity, improve mood or memory, lower blood cholesterol or blood sugar levels, fight inflammation, remove toxins, prevent or cure a disease, make your hair/nails/skin/digestion better, slow aging… Claims about a particular ingredient in foods/beverages that’s supposed to be “bad,” “toxic,” or contribute to a particular health problem (acne, autism, ADHD, PCOS, diabetes, cancer, Alzheimer’s disease, aging, hormone disruption, infertility, obesity, digestive problems)… Sources of Nutrition Claims Google! Magazines, Newspapers, Blogs Books, Videos, Documentaries Advertisements, Social Media Influencers Product label, brochure, website Scientific peer-reviewed journals
  • 2. How to choose a claim… Examples: Magazine article or blog claiming Intermittent fasting or whole 30 or keto is the answer to weight loss. Vitamin D/C/zinc and COVID19. LA Times article reporting on a new study that shows chocolate or red wine protects the heart (in time for Valentine’s Day) Book or Youtube video that claims sugar or wheat or gluten is toxic Documentary that claims plant-based diet best for performance (The Game Changers) Advertisement about new dietary supplement or “cleanse” for brain health, skin health, digestive health (turmeric, collagen, probiotics, spirulina, apple cider vinegar) Website bodybuilding.com claiming need certain amount/type of protein to get huge muscles. Or no soy/no dairy for PCOS or fertility. 2 Evaluating Nutrition Research & Claims Is the source credible & unbiased? Author/credentials “Nutritionists” vs. “Registered Dietitians” – what’s the difference? Self-proclaimed guru, fitness trainer, massage therapist, store clerk MDs, DCs, PhDs – are they always reliable? Is there any conflict of interest? Are they trying to sell you something? Publication source Internet site (.com or .org, .edu, .gov) Magazine, newsletter, brochure, trade journal (paid advertising) Peer-reviewed, professional/scientific journal
  • 3. Most “nutritionists” have little to no formal education/degree (e.g. famous people, fitness trainer/massage therapist/GNC or health food store clerk). Some “nutritionists” do have a high level of education/degree, but they may or may not be highly educated in nutrition Conflict of interest – Juice plus, herbal life, arbonne sales rep directly trying to sell you something or researcher/author/speaker could be employed/paid by the company trying to sell something (funded by beef/dairy council) Example of ephedra article in fitness magazine, local SCV magazines 3 Evaluating Nutrition Research & Claims How good is the research? Study design No systematic method at all testimonials, anecdotal, before/after Epidemiological / Observational Studies only show correlations, NOT cause and effect (due to confounding variables) Intervention / Experimental Studies CAN show cause & effect (because confounding variables are controlled) Best if study 1) randomized, 2) placebo-controlled, AND 3) double-blinded # of subjects / type of subjects Study length Especially when looking at long term weight loss and health/safety
  • 4. Epidemiological: Look at population of people’s lifestyle habits and look for associations/correlations with health/disease outcomes. Ex. Study shows eating ice cream is associated with drowning (that doesn’t show eating ice cream causes drowning…what confounding factor might be the reason for the association? People tend to eat more ice cream in the summer. People also tend to swim more in the summer.) Ex. Poor sperm quality linked to more phone/laptop use at night. Confounding factor is less quality sleep linked with both. Ex: People who drink red wine have less heart disease (based on Mediterranean diet, but could be less sat fat red meat, more olive oil MUFAs, more fruits/vegs, more active lifestyle, and/or genetics/ethnicity) Ex: Americans eating more carbs and gaining weight (but americans also eating more calories!) Intervention: Give a group of people an intervention/treatment and then see what effect it has on health/disease outcome. Ex: Hydroxycut/diet pill 8 week placebo-controlled study with same diet/exercise (but not randomized) Caffeine revs metabolism initially but effects wear off, no effect on weight/body fat loss. Long term keto diet effects? Subjects: Animals/rats vs. humans., Post menopausal women vs. young men (soy), congestive heart failure pts. Vs. young men (arginine/nitric oxide supplements) 4
  • 5. Be cautious with Observational Studies Observational studies do NOT prove one thing causes the other. They only show that two things are associated with one another. The REAL cause may be due to a confounding variable that is associated with both things. NOTE: In this example, poor quality sleep is the confounding variable since it results from late night electronics and is the real cause of poor sperm quality. Evaluating Nutrition Research & Claims Are the findings put in perspective? How many studies show a positive vs. negative or “null” effect? Any warning about adverse side effects/risks? Is the effective “dose” the same as what’s commonly consumed in a food or supplement? Any acknowledgement of other factors that have a more significant impact on the outcome than the one being studied? e.g. Risk of getting cancer from NOT eating vegetables is far greater than the cancer risk from ingesting trace amounts of pesticide residues on those vegetables. # studies: Anyone can “cherry-pick” studies to find only those 2 that support the claim (without mentioning the hundreds that don’t)
  • 6. Safety: Keto and intermittent fasting in certain populations harmful Reasonable dose: omega 3s DHA added to foods Physiologically significant: pesticide resides may be higher in conventional than organic, but the cancer risk of NOT eating any fruits and veggies is far greater 6 Using Library Resources www.aging-us.com 2764 AGING INTRODUCTION Major depressive disorder (MDD) is viewed as a major public health problem globally. MDD has a substantial impact on society and individuals, such as increasing economic burden and decreasing labor productivity [1–3]. At a global level, more than 300 million people are estimated to suffer from MDD, which is equivalent
  • 7. to 4.4% of the world’s population [4]. However, the pathogenesis of MDD is still unclear. Some theories have been developed to explain the biological mechanisms of MDD, such as neurotrophic alterations www.aging-us.com AGING 2020, Vol. 12, No. 3 Research Paper Age-specific differential changes on gut microbiota composition in patients with major depressive disorder Jian-Jun Chen1,2,*,#, Sirong He3,*, Liang Fang2,4,*, Bin Wang1, Shun-Jie Bai5, Jing Xie6, Chan-Juan Zhou7, Wei Wang8, Peng Xie4,7,8,# 1Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China 2Chongqing Key Laboratory of Cerebral Vascular Disease Research, Chongqing Medical University, Chongqing 400016, China 3Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China 4Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China 5Department of Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China 6Department of Endocrinology and Nephrology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, China 7NHC Key Laboratory of Diagnosis and Treatment on Brain
  • 8. Functional Diseases, Chongqing Medical University, Chongqing 400016, China 8Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China *Equal contribution #Co-senior authors Correspondence to: Peng Xie, Jian-Jun Chen; email: [email protected], [email protected] Keywords: major depressive disorder, gut microbiota, Firmicutes, Bacteroidetes, Actinobacteria Received: November 21, 2019 Accepted: January 12, 2020 Published: February 10, 2020 Copyright: Chen et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Emerging evidence has shown the age-related changes in gut microbiota, but few studies were conducted to explore the effects of age on the gut microbiota in patients with major depressive disorder (MDD). This study was performed to identify the age-specific differential gut microbiota in MDD patients. In total, 70 MDD patients and 71 healthy controls (HCs) were recruited and divided into two groups: young group (age 18-29 years) and middle-aged group (age 30-59 years). The 16S rRNA gene sequences were extracted from the collected fecal samples. Finally, we
  • 9. found that the relative abundances of Firmicutes and Bacteroidetes were significantly decreased and increased, respectively, in young MDD patients as compared with young HCs, and the relative abundances of Bacteroidetes and Actinobacteria were significantly decreased and increased, respectively, in middle-aged MDD patients as compared with middle-aged HCs. Meanwhile, six and 25 differentially abundant bacterial taxa responsible for the differences between MDD patients (young and middle-aged, respectively) and their respective HCs were identified. Our results demonstrated that there were age-specific differential changes on gut microbiota composition in patients with MDD. Our findings would provide a novel perspective to uncover the pathogenesis underlying MDD. mailto:[email protected] mailto:[email protected] www.aging-us.com 2765 AGING and neurotransmission deficiency [5, 6]. However, none of these theories has been universally accepted. Therefore, there is a pressing need to identify novel pathophysiologic mechanisms underlying this disease. In recent years, mounting evidence has shown that gut microbiota could play a vital role in every aspect of physiology [7]. It is the largest and most direct external
  • 10. environment of humans. Previous studies found that the disturbance of gut microbiota had a crucial role in the pathogenesis of many diseases [8–10]. Recent studies reported that gut microbiota could affect the host brain function and host behaviors through microbiota-gut- brain axis [11, 12]. Using germ-free mice, we found that gut microbiota could influence the gene levels in the hippocampus of mice and lipid metabolism in the prefrontal cortex of mice [13, 14]. Our clinical studies demonstrated that the disturbance of gut microbiota might be a contributory factor in the development of MDD [15, 16]. Nowadays, emerging evidence has shown the age- related changes in gut microbiota composition. For example, Firmicutes is the dominant taxa during the neonatal period, but Actinobacteria and Proteobacteria are about to increase in three to six months [17]. While
  • 11. in adults, Vemuri et al. reported that Bacteroidetes and Firmicutes were the dominant taxa [18]. Meanwhile, compared to younger individuals, the abundance of Bacteroidetes is significantly higher in frailer older individuals [19]. These results showed that there was a close relationship between age and gut microbiota composition. Ignoring this relationship would affect the robust of results when exploring the mechanism of action of gut microbiota in diseases. Therefore, to study the relationship between gut microbiota and MDD patients in different age groups, we recruited 52 young subjects aged from 18 to 29 years (27 healthy controls (HCs) and 25 MDD patients) and 89 middle-aged subjects aged from 30 to 59 years (44 HCs and 45 MDD patients). The main purpose of this study was to identify the age-specific differential changes on gut microbiota composition in MDD patients. Our results would display the different changes of gut microbiota
  • 12. composition along with age between HCs and MDD patients. RESULTS Differential gut microbiota composition As shown in Figure 1, the results of abundance-based coverage estimator (ACE) and Chao1 showed that there was no significant difference in OTU richness between MDD patients (young and middle-aged, respectively) and their respective HCs. However, the OPLS-DA model built with young HCs and young MDD patients showed an obvious difference in microbial abundances between these two groups (Figure 2A). The relative abundances of Firmicutes and Bacteroidetes were Figure 1. Comparison of alpha diversity between HCs and MDD patients. (A, B) ACE and Chao1 indexes showed no significant differences between young HCs (n=27) and young MDD patients (n=25); (C, D) ACE and Chao1 indexes showed no significant differences
  • 13. between middle-aged HCs (n=44) and middle-aged MDD patients (n=45). www.aging-us.com 2766 AGING significantly decreased and increased, respectively, in young MDD patients as compared with young HCs (Figure 2B). Meanwhile, the OPLS-DA model built with middle-aged HCs and middle-aged MDD patients showed an obvious difference in microbial abundances between these two groups (Figure 3A). The relative abundances of Bacteroidetes and Actinobacteria were significantly decreased and increased, respectively, in middle-aged MDD patients as compared with middle- aged HCs (Figure 3B). Key discriminatory OTUs In order to find out the gut microbiota primarily responsible for the separation between MDD patients (young and middle-aged, respectively) and their
  • 14. respective HCs, the Random Forests classifier was used. A total of 92 OTUs responsible for the separation between young MDD patients and young HCs were identified (Figure 4). These OTUs were mainly assigned to the Families of Bacteroidaceae, Clostridiaceae_1, Figure 2. 16S rRNA gene sequencing reveals changes to microbial abundances in young MDD patients. (A) OPLS-DA model showed an obvious difference in microbial abundances between the two groups (HCs, n=27; MDD, (n=25); (B) the relative abundances of Firmicutes and Bacteroidetes were significantly changed in young MDD patients (n=25) as compared with young HCs (n=27). Figure 3. 16S rRNA gene sequencing reveals changes to microbial abundances in middle-aged MDD patients. (A) OPLS- DA model showed an obvious difference in microbial abundances between the two groups (HCs, n=44; MDD, (n=45); (B) the relative abundances of Bacteroidetes and Actinobacteria were significantly changed in middle-aged MDD patients (n=45) as compared with middle- aged HCs (n=44).
  • 15. www.aging-us.com 2767 AGING Coriobacteriaceae, Erysipelotrichaceae, Lachnospiraceae, Peptostreptococcaceae and Ruminococcaceae. Meanwhile, a total of 122 OTUs responsible for the separation between middle-aged MDD patients and middle-aged HCs were identified (Figure 5). These OTUs were mainly assigned to the Families of Lachnospiraceae, Coriobacteriaceae, Streptococcaceae, Prevotellaceae, Bacteroidaceae, Eubacteriaceae, Actinomycetaceae, Sutterellaceae, Acidaminococcaceae, Erysipelotrichaceae, Ruminococcaceae, and Porphyromonadaceae. Figure 4. Heatmap of discriminative OTUs abundances between young HCs (n=27) and young MDD patients (n=25). Figure 5. Heatmap of discriminative OTUs abundances between middle-aged HCs (n=44) and middle-aged MDD patients (n=45).
  • 16. www.aging-us.com 2768 AGING Differentially abundant bacterial taxa Differentially abundant bacterial taxa responsible for the differences between MDD patients (young and middle-aged, respectively) and their respective HCs were identified by the metagenomic Linear Discriminant Analysis (LDA) Effect Size (LEfSe) approach (LDA score>2.0 and p-value<0.05). In total, six bacterial taxa with statistically significant and biologically consistent differences in young MDD patients were identified (Figure 6). Meanwhile, fifteen bacterial taxa with statistically significant and biologically consistent differences in middle-aged MDD patients were identified (Figure 7). In addition, using Figure 6. Differentially abundant features identified by LEfSe that characterize significant differences between young HCs
  • 17. (n=27) and young MDD patients (n=25). Figure 7. Differentially abundant features identified by LEfSe that characterize significant differences between middle-aged HCs (n=44) and middle-aged MDD patients (n=45). www.aging-us.com 2769 AGING the receiver operating characteristic (ROC) curve analysis, we found that Clostridium_sensu_stricto, Clostridium_XI and Clostridium_XVIII showed good diagnostic performance (area under the curve (AUC) >0.7) in diagnosing young MDD patients (Figure 8A– 8C). We also found that Anaerostipes, Streptococcus, Blautia, Faecalibacterium and Roseburia showed good diagnostic performance (AUC>0.7) in diagnosing middle-aged MDD patients (Figure 8D–8H). Effects of age on microbial abundances Using the LEfSe approach, we identified four
  • 18. differentially abundant bacterial taxa (the Family level) between young HCs and middle-aged HCs (Streptococcaceae, Coriobacteriaceae, Carnobacteriaceae and Clostridiales_Incertae_Sedis_XIII) (Figure 9A); we also identified six differentially abundant bacterial taxa (the Family level) between young MDD patients and middle-aged MDD patients (Prevotellaceae, Acidaminococcaceae, Veillonellaceae Peptostrep- tococcaceae, Lachnospiraceae and Ruminococcaceae) (Figure 9B). Meanwhile, using the LEfSe approach, we identified five differentially abundant bacterial taxa (the Genus level) between young HCs and middle-aged HCs (Streptococcus, Veillonella, Granulicatella, Collinsella and Megamonas) (Figure 10A). All these bacterial taxa were significantly decreased in middle-aged HCs; we also identified nine differentially abundant bacterial taxa (the Genus level) between young MDD patients and middle-aged MDD patients (Megamonas,
  • 19. Prevotella, Phascolarctobacterium, Anaerostipes, Clostridium_XVIII, Gordonibacter, Eggerthella, Clostridium_XI and Turicibacter) (Figure 10B). Effects of medication on microbial abundances To determinate the homogeneity of gut microbiota composition between medicated and non-medicated MDD patients, we firstly used the middle-aged HCs and non-medicated middle-aged MDD patients to built OPLS-DA model (Figure 11A). The results showed that 41 of the 44 middle-aged HCs and 30 of the 31 non- medicated middle-aged MDD patients were correctly diagnosed by the OPLS-DA model. Then, we used the built model to predict class membership of 14 medicated middle-aged MDD patients. The T-predicted scatter plot showed that 11 of the 14 medicated middle- aged MDD patients were correctly predicted (Figure 11B). These finding indicated that the gut microbiota
  • 20. composition of non-medicated middle-aged MDD patients were distinct from middle-aged HCs, but not from medicated middle-aged MDD patients. DISCUSSION Individuals in the different phases of life cycle (named children, young, middle-aged and elderly) present different biological characteristics and disease risks [20]. Understanding the different characteristics of patients in particular age phases could be facilitated to prevent and treat diseases. According to the World Health Organization reported, the prevalence rates of depression vary by age, peaking in older adulthood. It also occurs in children, but at a lower level compared with older age groups. Here, we conducted this work to investigate how the gut microbiota composition changed in different age phases of MDD patients, and found some age-specific differential gut microbiota in
  • 21. Figure 8. Differential taxa (at the genus level) with AUC>0.7 in diagnosing MDD patients from HCs. (A–C) the diagnostic performances of three taxa in diagnosing young MDD patients (n=25) from young HCs (n=27); (D–H) the diagnostic performances of five taxa in diagnosing middle-aged MDD patients (n=45) from middle- aged HCs (n=44). www.aging-us.com 2770 AGING Figure 9. 16S rRNA gene sequencing reveals changes to microbial abundances at family level (Mean±SEM). (A) the abundances of four taxonomic levels were significantly changed between young HCs (n=27) and middle-aged HCs (n=44); (B) the abundances of six taxonomic levels were significantly changed between young MDD patients (n=25) and middle-aged MDD patients (n=45). Figure 10. 16S rRNA gene sequencing reveals changes to microbial abundances at genus level (Mean±SEM). (A) the abundances of five taxonomic levels were significantly changed between young HCs (n=27) and middle-aged HCs (n=44); (B) the abundances of nine taxonomic levels were significantly changed between young MDD patients (n=25) and middle-aged MDD patients (n=45).
  • 22. www.aging-us.com 2771 AGING MDD patients. Our results could provide a new perspective on exploring the pathogenesis of MDD. Many previous studies focused on the effects of gut microbiota on brain functions [21, 22]. However, few studies have taken the effects of age on gut microbiota into consideration when exploring the pathogenesis of MDD. Our previous study found that the relative abundances of Bacteroidetes and Actinobacteria were significantly decreased and increased, respectively, in MDD patients as compared with HCs [15]. But, in this study, we found that the relative abundances of Firmicutes and Bacteroidetes were significantly decreased and increased, respectively, in young MDD patients as compared with young HCs, and the relative abundances of Bacteroidetes and Actinobacteria were
  • 23. significantly decreased and increased, respectively, in middle-aged MDD patients as compared with middle- aged HCs. This disparity might be caused by the different age structures. Meanwhile, only 35 key discriminatory OTUs were significantly changed in both young (92 key discriminatory OTUs) and middle-aged (127 key discriminatory OTUs) MDD patients. Moreover, the differentially abundant bacterial taxa in young and middle-aged MDD patients were totally different at both Family level and Genus level. These results demonstrated that it was necessary to identify the age-specific differential gut microbiota in patients with MDD. As far as we known, gut microbiota composition and its function could be easily influenced by many factor, such as gender, age, life experiences, dietary habit and genetics. Mariat et al reported that the
  • 24. Firmicutes/Bacteroidetes ratio of the human microbiota could change with age [23]. Interestingly, here we found that the relative abundance of Firmicutes was significantly decreased in young MDD patients, but not in middle-aged MDD patients; the relative abundance of Bacteroidetes was significantly increased and decreased, respectively, in young and middle-aged MDD patients. In our previous studies, we did not analyze the potential effects of medication on gut microbiota composition in MDD patients [15, 16]. Here, due to the small samples of young group, we only used the middle-aged group to analyze the effects of Figure 11. Assessment of gut microbiota composition in non- medicated and medicated middle-aged MDD patients. (A) middle-aged HCs (n=44) and non-medicated middle-aged MDD patients (n=31) were effectively separated by the built OPLS- DA model; (B) 14 medicated middle-aged MDD patients were correctly predicted by the model.
  • 25. www.aging-us.com 2772 AGING medication on the gut microbiota composition. The results showed that the medication seemed to have little effects on gut microbiota composition in MDD patients. However, our findings had to be cautiously interpreted due to the relatively small samples using to analyze the effects of medication on gut microbiota composition. The relative abundance of genus Clostridium_XVIII was not found to be significantly different between MDD patients and HCs in our previous study [15]. However, in this study, we found that the relative abundance of genus Clostridium_XVIII was significantly decreased in young MDD patients compared with young HCs, while increased in middle- aged MDD patients compared with middle-aged HCs. The reason of this disparity might be that age could significantly affect the relative abundance of genus
  • 26. Clostridium_XVIII in MDD patients, but not HCs: i) compared to young MDD patients, the middle-aged MDD patients had a significantly higher relative abundance of genus Clostridium_XVIII; and ii) the relative abundance of genus Clostridium_XVIII was similar between young and middle-aged HCs. Meanwhile, we found that the relative abundance of genus Megamonas was significantly decreased in both middle-aged HCs and middle-aged MDD patients compared to their respective young populations. In addition, most of differential bacterial taxa were significantly decreased in middle-aged HCs compared with young HCs, but only about half of differential bacterial taxa were significantly decreased in middle- aged MDD patients compared with young MDD patients. Lozupone et al. reported that gut microbiota could not only simply determine the certain host characteristics, but also respond to signals from host via
  • 27. multiple feedback loops [24]. Therefore, our results suggested that age might have the different effects on the gut microbiota composition of HCs and MDD patients, and should always be considered in investigating the relationship between MDD and gut microbiota. Limitations should be mentioned here. Firstly, the number of HCs and MDD patients was relatively small, and future works were still needed to further study and support our results. Secondly, we only explored the age- specific differential changes on gut microbiota composition in patients with MDD; future studies should further investigate the functions of these identified differential gut microbiota using metagenomic technology. Thirdly, all included subjects were from the same site and ethnicity; thus, the potential site- and ethnic-specific biases in microbial phenotypes could not be ruled out, which might limit
  • 28. the applicability of our results [25–28]. Fourthly, only young and middle-aged groups were recruited, future studies should recruit old-aged group and children group to further identify the age-specific differential gut microbiota in the different phases of life cycle. Fifthly, we only investigated the differences in gut microbiota between HCs and MDD patients on phylum level, family level and genus level. Future studies were needed to further explore the differences on other levels, such as class level and species level. Sixthly, we did not collect information on smoking, a factor which could influence the gut microbiota composition. Future studies were needed to analyze how the smoking influenced the gut microbiota composition in the different phases of life cycle of subjects. Finally, we found that the medication status of subjects could not significantly affect our results. However, limited by the relatively small samples, this conclusion was needed
  • 29. future studies to further validate. In conclusion, in this study, we found that there were age-specific differential changes on gut microbiota composition in patients with MDD, and identified some age-specific differentially abundant bacterial taxa in MDD patients. Our findings would provide a novel perspective to uncover the pathogenesis underlying MDD, and potential gut-mediated therapies for MDD patients. Limited by the small number of subjects, the results of the present study were needed future studies to validate and support. MATERIALS AND METHODS Subject recruitment This study was approved by the Ethical Committee of Chongqing Medical University and conformed to the provisions of the Declaration of Helsinki. In total, there
  • 30. were 27 young HCs (aged 18-29 years) and 25 young MDD outpatients (aged 18-29 years) in the young group; there were 44 middle-aged HCs (aged 30-59 years) and 45 middle-aged MDD outpatients (aged 30- 59 years) in the middle-aged group. Most of MDD patients were first-episode drug-naïve depressed subjects. There were only seven young MDD patients and 14 middle-aged MDD patients receiving medications. The detailed information of these included subjects was described in Table 1. All HCs were recruited from the Medical Examination Center of Chongqing Medical University, and all MDD patients were recruited from the psychiatric center of Chongqing Medical University. MDD patients were screened in the baseline interview by two experienced psychiatrists using the DSM-IV (Diagnostic and Statistical Manual of Mental Disorders, 4th Edition)-based Composite International Diagnostic Interview (CIDI, version2.1).
  • 31. The Hamilton Depression Rating Scale (HDRS) was used to assess the depressive symptoms of each patient, www.aging-us.com 2773 AGING Table 1. Demographic and clinical characteristics of MDD patients and HCsa. Young group (18-29 years) Middle-aged group (30-59 years) HC MDD p-value HC MDD p-value Sample Size 27 25 – 44 45 – Age (years)c 24.96±2.31 24.0±3.74 0.26 47.16±8.07 44.96±7.76 0.19 Sex (female/male) 19/8 18/7 0.89 34/10 31/14 0.37 BMI 21.53±2.37 22.13±2.24 0.35 23.23±2.33 22.64±2.64 0.26 Medication (Y/N) 0/27 7/18 – 0/44 14/31 – HDRS scores 0.29±0.61 22.64±3.18 <0.00001 0.34±0.74 23.0±4.61 <0.00001 aAbbreviations: HDRS: Hamilton Depression Rating Scale; HCs: healthy controls; MDD: major depressive disorder; BMI: body mass index.
  • 32. and those patients with HDRS score >=17 were included. Meanwhile, MDD patients were excluded if they had other mental disorders, illicit drug use or substance abuse, and were pregnant or menstrual women. HCs were excluded if they were with mental disorders, illicit drug use or systemic medical illness. All the included subjects provided written informed consent before sample collection. 16s rRNA gene sequencing We used the standard PowerSoil kit protocol to extract the bacterial genomic DNA from the fecal samples. Briefly, we thawed the frozen fecal samples on ice and pulverized the samples with a pestle and mortar in liquid nitrogen. After adding MoBio lysis buffer into the samples and mixing them, the suspensions were centrifuged. The obtained supernatant was moved into the MoBio Garnet bead tubes containing MoBio buffer.
  • 33. Subsequently, we used the Roche 454 sequencing (454 Life Sciences Roche, Branford, PA, USA) to extract the bacterial genomic DNA. The extracted V3-V5 regions of 16S rRNA gene were polymerase chain reaction- amplified with bar-coded universal primers containing linker sequences for pyrosequencing [29]. The Mothur 1.31.2 (http://www.mothur.org/) was used to quality-filtered the obtained raw sequences to identify unique reads [30]. Raw sequences met any one of the following criteria were excluded: i) less than 200bp or greater than 1000bp; ii) contained any ambiguous bases, primer mismatches, or barcode mismatches; and iii) homopolymer runs exceeding six bases. The remaining sequences were assigned to operational taxonomic units (OTUs) with 97% threshold, and then taxonomically classified according to Ribosomal Database Project (RDP) reference
  • 34. database [31]. We used these taxonomies to construct the summaries of the taxonomic distributions of OTUs, and then calculated the relative abundances of gut microbiota at different levels. The abovementioned procedure and most of data were from our previous studies [15, 16]. Statistical analysis Richness was one of the two most commonly used alpha diversity measurements. Here, we used two different parameters (Chao1 and ACE) to estimate the OTU richness [32, 33]. The orthogonal partial least squares discriminant analysis (OPLS-DA) was a multivariate method, which was used to remove extraneous variance (unrelated to the group) from the sequencing datasets. The LEfSe was a new analytical method for discovering the metagenomic biomarker by class comparison. The bacterial taxa with LDA score>2.0 were viewed as the
  • 35. differentially abundant bacterial taxa responsible for the differences between different groups. Here, both OPLS- DA [34, 35] and LEfSe were used to reduce the dimensionality of datasets and identify the differentially abundant bacterial taxa (the Family level and Genus level) that could be used to characterize the significant differences between HCs and MDD patients. Meanwhile, we used the Random Forest algorithm to identify the critical discriminatory OTUs. The ROC curve analysis was used to assess the diagnostic performance of these identified differential bacterial taxa. The AUC was the evaluation index. Finally, we used the LEfSe to reveal the changes of microbial abundances at Family level and Genus level in HCs and MDD patients, respectively. ACKNOWLEDGMENTS Our sincere gratitude is extended to Professors Delan Yang and Hua Hu from Psychiatric Center of the First
  • 36. Affiliated Hospital of Chongqing Medical University for their efforts in sample collection. CONFLICTS OF INTEREST The authors declare no financial or other conflicts of interest. http://www.mothur.org/ www.aging-us.com 2774 AGING FUNDING This work was supported by the National Key R&D Program of China (2017YFA0505700), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019PT320002300), the Natural Science Foundation Project of China (81820108015, 81701360, 81601208, 81601207), the Chongqing Science and Technology Commission (cstc2017jcyjAX0377), the Chongqing Yuzhong
  • 37. District Science and Technology Commission (20190115), and supported by the fund from the Joint International Research Laboratory of Reproduction & Development, Institute of Life Sciences, Chongqing Medical University, Chongqing, China, and also supported by the Scientific Research and Innovation Experiment Project of Chongqing Medical University (CXSY201862, CXSY201863). REFERENCES 1. Yirmiya R, Rimmerman N, Reshef R. Depression as a microglial disease. Trends Neurosci. 2015; 38:637–58. https://doi.org/10.1016/j.tins.2015.08.001 PMID:26442697 2. Pan JX, Xia JJ, Deng FL, Liang WW, Wu J, Yin BM, Dong MX, Chen JJ, Ye F, Wang HY, Zheng P, Xie P. Diagnosis of major depressive disorder based on changes in multiple plasma neurotransmitters: a targeted metabolomics study. Transl Psychiatry. 2018; 8:130. https://doi.org/10.1038/s41398-018-0183-x PMID:29991685
  • 38. 3. Zhao H, Du H, Liu M, Gao S, Li N, Chao Y, Li R, Chen W, Lou Z, Dong X. Integrative proteomics–metabolomics strategy for pathological mechanism of vascular depression mouse model. J Proteome Res. 2018; 17:656–69. https://doi.org/10.1021/acs.jproteome.7b00724 PMID:29190102 4. Stringaris A. Editorial: what is depression? J Child Psychol Psychiatry. 2017; 58:1287–89. https://doi.org/10.1111/jcpp.12844 PMID:29148049 5. Luscher B, Shen Q, Sahir N. The GABAergic deficit hypothesis of major depressive disorder. Mol Psychiatry. 2011; 16:383–406. https://doi.org/10.1038/mp.2010.120 PMID:21079608 6. Guilloux JP, Douillard-Guilloux G, Kota R, Wang X, Gardier AM, Martinowich K, Tseng GC, Lewis DA, Sibille E. Molecular evidence for BDNF- and GABA-related dysfunctions in the amygdala of female subjects with major depression. Mol Psychiatry. 2012; 17:1130–42. https://doi.org/10.1038/mp.2011.113 PMID:21912391 7. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012; 148:1258–70. https://doi.org/10.1016/j.cell.2012.01.035 PMID:22424233
  • 39. 8. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, Camporez JP, Shulman GI, Gordon JI, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 2012; 482:179–85. https://doi.org/10.1038/nature10809 PMID:22297845 9. Peng W, Yi P, Yang J, Xu P, Wang Y, Zhang Z, Huang S, Wang Z, Zhang C. Association of gut microbiota composition and function with a senescence- accelerated mouse model of Alzheimer’s Disease using 16S rRNA gene and metagenomic sequencing analysis. Aging (Albany NY). 2018; 10:4054–65. https://doi.org/10.18632/aging.101693 PMID:30562162 10. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA, Gordon JI, Chervonsky AV. Innate immunity and intestinal microbiota in the development of Type 1 diabetes. Nature. 2008; 455:1109–13. https://doi.org/10.1038/nature07336 PMID:18806780 11. Tillisch K. The effects of gut microbiota on CNS function in humans. Gut Microbes. 2014; 5:404–10. https://doi.org/10.4161/gmic.29232 PMID:24838095 12. Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, Deng Y, Blennerhassett P, Macri J, McCoy KD, Verdu EF, Collins SM. The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior
  • 40. in mice. Gastroenterology. 2011; 141:599–609, 609.e1–3. https://doi.org/10.1053/j.gastro.2011.04.052 PMID:21683077 13. Chen JJ, Zeng BH, Li WW, Zhou CJ, Fan SH, Cheng K, Zeng L, Zheng P, Fang L, Wei H, Xie P. Effects of gut microbiota on the microRNA and mRNA expression in the hippocampus of mice. Behav Brain Res. 2017; 322:34–41. https://doi.org/10.1016/j.bbr.2017.01.021 PMID:28093256 14. Chen JJ, Xie J, Zeng BH, Li WW, Bai SJ, Zhou C, Chen W, Wei H, Xie P. Absence of gut microbiota affects lipid metabolism in the prefrontal cortex of mice. Neurol Res. 2019; 41:1104–12. https://doi.org/10.1080/01616412.2019.1675021 PMID:31587617 15. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, Chen J, Fan S, Du X, Zhang X, Yang D, Yang Y, et al. Gut https://doi.org/10.1016/j.tins.2015.08.001 https://www.ncbi.nlm.nih.gov/pubmed/26442697 https://doi.org/10.1038/s41398-018-0183-x https://www.ncbi.nlm.nih.gov/pubmed/29991685 https://doi.org/10.1021/acs.jproteome.7b00724 https://www.ncbi.nlm.nih.gov/pubmed/29190102 https://doi.org/10.1111/jcpp.12844 https://www.ncbi.nlm.nih.gov/pubmed/29148049 https://doi.org/10.1038/mp.2010.120 https://www.ncbi.nlm.nih.gov/pubmed/21079608
  • 41. https://doi.org/10.1038/mp.2011.113 https://www.ncbi.nlm.nih.gov/pubmed/21912391 https://doi.org/10.1016/j.cell.2012.01.035 https://www.ncbi.nlm.nih.gov/pubmed/22424233 https://doi.org/10.1038/nature10809 https://www.ncbi.nlm.nih.gov/pubmed/22297845 https://doi.org/10.18632/aging.101693 https://www.ncbi.nlm.nih.gov/pubmed/30562162 https://doi.org/10.1038/nature07336 https://www.ncbi.nlm.nih.gov/pubmed/18806780 https://doi.org/10.4161/gmic.29232 https://www.ncbi.nlm.nih.gov/pubmed/24838095 https://doi.org/10.1053/j.gastro.2011.04.052 https://www.ncbi.nlm.nih.gov/pubmed/21683077 https://doi.org/10.1016/j.bbr.2017.01.021 https://www.ncbi.nlm.nih.gov/pubmed/28093256 https://doi.org/10.1080/01616412.2019.1675021 https://www.ncbi.nlm.nih.gov/pubmed/31587617 www.aging-us.com 2775 AGING microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol Psychiatry. 2016; 21:786–96. https://doi.org/10.1038/mp.2016.44 PMID:27067014 16. Chen JJ, Zheng P, Liu YY, Zhong XG, Wang HY, Guo YJ, Xie P. Sex differences in gut microbiota in patients with major depressive disorder. Neuropsychiatr Dis Treat. 2018; 14:647–55. https://doi.org/10.2147/NDT.S159322 PMID:29520144
  • 42. 17. Lim ES, Zhou Y, Zhao G, Bauer IK, Droit L, Ndao IM, Warner BB, Tarr PI, Wang D, Holtz LR. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med. 2015; 21:1228–34. https://doi.org/10.1038/nm.3950 PMID:26366711 18. Vemuri R, Gundamaraju R, Shastri MD, Shukla SD, Kalpurath K, Ball M, Tristram S, Shankar EM, Ahuja K, Eri R. Gut Microbial Changes, Interactions, and Their Implications on Human Lifecycle: An Ageing Perspective. Biomed Res Int. 2018; 2018:4178607. https://doi.org/10.1155/2018/4178607 PMID:29682542 19. Claesson MJ, Cusack S, O’Sullivan O, Greene-Diniz R, de Weerd H, Flannery E, Marchesi JR, Falush D, Dinan T, Fitzgerald G, Stanton C, van Sinderen D, O’Connor M, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci USA. 2011 (Suppl 1); 108:4586–91. https://doi.org/10.1073/pnas.1000097107 PMID:20571116 20. Chia CW, Egan JM, Ferrucci L. Age-Related Changes in Glucose Metabolism, Hyperglycemia, and Cardiovascular Risk. Circ Res. 2018; 123:886–904. https://doi.org/10.1161/CIRCRESAHA.118.312806 PMID:30355075 21. Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012; 13:701–12.
  • 43. https://doi.org/10.1038/nrn3346 PMID:22968153 22. Gareau MG, Wine E, Rodrigues DM, Cho JH, Whary MT, Philpott DJ, Macqueen G, Sherman PM. Bacterial infection causes stress-induced memory dysfunction in mice. Gut. 2011; 60:307–17. https://doi.org/10.1136/gut.2009.202515 PMID:20966022 23. Mariat D, Firmesse O, Levenez F, Guimarăes V, Sokol H, Doré J, Corthier G, Furet JP. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009; 9:123. https://doi.org/10.1186/1471-2180-9-123 PMID:19508720 24. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012; 489:220–30. https://doi.org/10.1038/nature11550 PMID:22972295 25. Chen JJ, Bai SJ, Li WW, Zhou CJ, Zheng P, Fang L, Wang HY, Liu YY, Xie P. Urinary biomarker panel for diagnosing patients with depression and anxiety disorders. Transl Psychiatry. 2018; 8:192. https://doi.org/10.1038/s41398-018-0245-0 PMID:30232320 26. Hou L, Wei X, Zhuo Y, Qin L, Yang F, Zhang L, Song X.
  • 44. GC-MS-based metabolomics approach to diagnose depression in hepatitis B virus-infected patients with middle or old age. Aging (Albany NY). 2018; 10:2252–65. https://doi.org/10.18632/aging.101535 PMID:30178754 27. Chen JJ, Xie J, Zeng L, Zhou CJ, Zheng P, Xie P. Urinary metabolite signature in bipolar disorder patients during depressive episode. Aging (Albany NY). 2019; 11:1008–18. https://doi.org/10.18632/aging.101805 PMID:30721880 28. Chen JJ, Xie J, Li WW, Bai SJ, Wang W, Zheng P, Xie P. Age-specific urinary metabolite signatures and functions in patients with major depressive disorder. Aging (Albany NY). 2019; 11:6626–37. https://doi.org/10.18632/aging.102133 PMID:31493765 29. Tamaki H, Wright CL, Li X, Lin Q, Hwang C, Wang S, Thimmapuram J, Kamagata Y, Liu WT. Analysis of 16S rRNA amplicon sequencing options on the Roche/454 next-generation titanium sequencing platform. PLoS One. 2011; 6:e25263. https://doi.org/10.1371/journal.pone.002 5263 PMID:21966473 30. Yang S, Liebner S, Alawi M, Ebenhöh O, Wagner D. Taxonomic database and cut-off value for processing mcrA gene 454 pyrosequencing data by MOTHUR. J
  • 45. Microbiol Methods. 2014; 103:3–5. https://doi.org/10.1016/j.mimet.2014.05. 006 PMID:24858450 31. Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, Brown CT, Porras-Alfaro A, Kuske CR, Tiedje JM. Ribosomal Database Project: data and tools for high throughput rRNA analysis. Nucleic Acids Res. 2014; 42:D633–42. https://doi.org/10.1093/nar/gkt1244 PMID:24288368 32. Countway PD, Gast RJ, Dennett MR, Savai P, Rose JM, Caron DA. Distinct protistan assemblages characterize the euphotic zone and deep sea (2500 m) of the western North Atlantic (Sargasso Sea and Gulf Stream). https://doi.org/10.1038/mp.2016.44 https://www.ncbi.nlm.nih.gov/pubmed/27067014 https://doi.org/10.2147/NDT.S159322 https://www.ncbi.nlm.nih.gov/pubmed/29520144 https://doi.org/10.1038/nm.3950 https://www.ncbi.nlm.nih.gov/pubmed/26366711 https://doi.org/10.1155/2018/4178607 https://www.ncbi.nlm.nih.gov/pubmed/29682542 https://doi.org/10.1073/pnas.1000097107 https://www.ncbi.nlm.nih.gov/pubmed/20571116 https://doi.org/10.1161/CIRCRESAHA.118.312806 https://www.ncbi.nlm.nih.gov/pubmed/30355075 https://doi.org/10.1038/nrn3346 https://www.ncbi.nlm.nih.gov/pubmed/22968153 https://doi.org/10.1136/gut.2009.202515 https://www.ncbi.nlm.nih.gov/pubmed/20966022 https://doi.org/10.1186/1471-2180-9-123 https://www.ncbi.nlm.nih.gov/pubmed/19508720
  • 46. https://doi.org/10.1038/nature11550 https://www.ncbi.nlm.nih.gov/pubmed/22972295 https://doi.org/10.1038/s41398-018-0245-0 https://www.ncbi.nlm.nih.gov/pubmed/30232320 https://doi.org/10.18632/aging.101535 https://www.ncbi.nlm.nih.gov/pubmed/30178754 https://doi.org/10.18632/aging.101805 https://www.ncbi.nlm.nih.gov/pubmed/30721880 https://doi.org/10.18632/aging.102133 https://www.ncbi.nlm.nih.gov/pubmed/31493765 https://doi.org/10.1371/journal.pone.0025263 https://www.ncbi.nlm.nih.gov/pubmed/21966473 https://doi.org/10.1016/j.mimet.2014.05.006 https://www.ncbi.nlm.nih.gov/pubmed/24858450 https://doi.org/10.1093/nar/gkt1244 https://www.ncbi.nlm.nih.gov/pubmed/24288368 www.aging-us.com 2776 AGING Environ Microbiol. 2007; 9:1219–32. https://doi.org/10.1111/j.1462-2920.2007.01243.x PMID:17472636 33. Zuendorf A, Bunge J, Behnke A, Barger KJ, Stoeck T. Diversity estimates of microeukaryotes below the chemocline of the anoxic Mariager Fjord, Denmark. FEMS Microbiol Ecol. 2006; 58:476–91. https://doi.org/10.1111/j.1574-6941.2006.00171.x PMID:17117990 34. Shankar V, Agans R, Holmes B, Raymer M, Paliy O. Do gut microbial communities differ in pediatric IBS and
  • 47. health? Gut Microbes. 2013; 4:347–52. https://doi.org/10.4161/gmic.24827 PMID:23674073 35. Ramadan Z, Xu H, Laflamme D, Czarnecki-Maulden G, Li QJ, Labuda J, Bourqui B. Fecal microbiota of cats with naturally occurring chronic diarrhea assessed using 16S rRNA gene 454-pyrosequencing before and after dietary treatment. J Vet Intern Med. 2014; 28:59–65. https://doi.org/10.1111/jvim.12261 PMID:24592406 https://doi.org/10.1111/j.1462-2920.2007.01243.x https://www.ncbi.nlm.nih.gov/pubmed/17472636 https://doi.org/10.1111/j.1574-6941.2006.00171.x https://www.ncbi.nlm.nih.gov/pubmed/17117990 https://doi.org/10.4161/gmic.24827 https://www.ncbi.nlm.nih.gov/pubmed/23674073 https://doi.org/10.1111/jvim.12261 https://www.ncbi.nlm.nih.gov/pubmed/24592406 RESEARCH Open Access Associations between gut microbiota and Alzheimer’s disease, major depressive disorder, and schizophrenia Zhenhuang Zhuang1, Ruotong Yang1, Wenxiu Wang1, Lu Qi2,3* and Tao Huang1,4,5,6* Abstract
  • 48. Background: Growing evidence has shown that alterations in the gut microbiota composition were associated with a variety of neuropsychiatric conditions. However, whether such associations reflect causality remains unknown. We aimed to reveal the causal relationships among gut microbiota, metabolites, and neuropsychiatric disorders including Alzheimer’s disease (AD), major depressive disorder (MDD), and schizophrenia (SCZ). Methods: A two-sample bi-directional Mendelian randomization analysis was performed by using genetic variants from genome-wide association studies as instrumental variables for gut microbiota, metabolites, AD, MDD, and SCZ, respectively. Results: We found suggestive associations of host-genetic- driven increase in Blautia (OR, 0.88; 95%CI, 0.79–0.99; P = 0.028) and elevated γ-aminobutyric acid (GABA) (0.96; 0.92–1.00; P = 0.034), a downstream product of Blautia-dependent arginine metabolism, with a lower risk of AD. Genetically increased Enterobacteriaceae family and Enterobacteriales order were potentially associated with a higher risk of SCZ (1.09; 1.00–1.18; P = 0.048), while Gammaproteobacteria class (0.90; 0.83–0.98; P = 0.011) was related to a lower risk for SCZ. Gut production of serotonin was potentially associated with an increased risk of SCZ (1.07; 1.00–1.15; P = 0.047). Furthermore, genetically increased Bacilli class was related to a higher risk of MDD (1.07; 1.02–1.12; P = 0.010). In the other direction, neuropsychiatric disorders altered gut microbiota composition. Conclusions: These data for the first time provide evidence of potential causal links between gut microbiome and AD, MDD, and SCZ. GABA and serotonin may play an important role in gut microbiota-host crosstalk in AD and SCZ, respectively. Further investigations in
  • 49. understanding the underlying mechanisms of associations between gut microbiota and AD, MDD, and SCZ are required. Keywords: Gut microbiota, Neuropsychiatric disorder, Mendelian randomization, Genetic association, Causality Background The human intestine comprises a very complex group of gut microbiota, which influence the risk of neuropsychiatric disorders [1, 2]. Accumulating evidence has suggested that microbiota metabolites such as neurotransmitters and short-chain fatty acids (SCFAs) may play a central role in microbiota-host crosstalk that regulates the brain function and behavior [3, 4]. Therefore, to understand the mechan- ism of the gut-brain axis in neuropsychiatric disorders may have clinical benefits. Observational studies, most of case-control designs, have shown differences in the composition of the gut microbiota between healthy individuals and patients with neuropsychi- atric disorders such as Alzheimer’s disease (AD), major depression disorder (MDD), and schizophrenia (SCZ); © The Author(s). 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons
  • 50. licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. * Correspondence: [email protected]; [email protected] 2Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA 1Department of Epidemiology & Biostatistics, School of Public Health, Peking University, 38 Xueyuan Road, Beijing 100191, China Full list of author information is available at the end of the article Zhuang et al. Journal of Neuroinflammation (2020) 17:288 https://doi.org/10.1186/s12974-020-01961-8 http://crossmark.crossref.org/dialog/?doi=10.1186/s12974-020- 01961-8&domain=pdf http://orcid.org/0000-0002-0328-1368 http://creativecommons.org/licenses/by/4.0/ http://creativecommons.org/publicdomain/zero/1.0/ mailto:[email protected] mailto:[email protected] however, such associations substantially differed across studies [5–7]. Noteworthy, genome-based metabolic mod-
  • 51. eling of the human gut microbiota revealed that several genera have predictive capability to produce or consume neurotransmitters (called microbial neurotransmitters) such as γ-aminobutyric acid (GABA) and serotonin [8, 9], which have been consistently shown to played a key role in the regulation of brain function [10, 11]. A meta-analysis of 35 observational studies reported that increased GABA levels were associated with a lower risk of AD [12]. In addition, a previous study (n = 40) reported that plasma serotonin was lower and platelet serotonin was higher in SCZ patients compared with controls [13], while another study showed that lower platelet serotonin concentrations were associated with depressive symptoms of SCZ (n = 364) [14]. There is no doubt that these small observational studies were sus- ceptible to confounding bias and reverse causation. It is crucial to elucidate whether such associations reflect causal relations or spurious correlations due to bias. Mendelian randomization (MR), which overcomes the bias due to confounding and reverse causation above- mentioned, has been widely used to assess causal rela- tionships by exploiting genetic variants as instrumental variables of the exposure [15]. Recent genetic studies have demonstrated that the host genetic variants influ- ence the gut microbiota composition [16–18]. Thus, such findings allowed us to deploy an MR approach to infer the mutually causal relations of gut microbiota and metabolites with neuropsychiatric disorders. Therefore, we for the first time applied a two-sample bi-directional MR approach to detect causal relation- ships among gut microbiota, metabolites, and diverse forms of neuropsychiatric disorders including AD, SCZ, and MDD. Methods
  • 52. Study design overview We employed a two-sample bi-directional MR approach to investigate the causal relationships among gut micro- biota, metabolites, and AD, MDD, or SCZ using summary-level data from large genome-wide association studies (GWASs) for gut microbiota and AD, MDD, or SCZ. Ethical approval for each study included in the MR analysis can be found in the original articles [19–23]. Data sources and instruments Gut microbiota We leveraged summary statistics from a GWAS of gut microbiota conducted among two independent but geo- graphically matched cohorts of European ancestry (n = 1812) using 16S rRNA gene sequencing (Table 1) [19], which yielded a total of 38 and 374 identified phyla and genera respectively. The GWAS defined a “core measur- able microbiota” after removing rare bacteria and investigating associations between host genetic variants and specific bacterial traits, including 40 operational taxonomic units (OTUs) and 58 taxa ranging from the genus to the phylum level. Accordingly, the GWAS fur- ther identified 54 genome-wide significant associations involving 40 loci and 22 bacterial traits (meta-analysis P < 5 × 10−8). We selected single nucleotide polymor- phisms (SNPs) at thresholds for genome-wide signifi- cance (P < 5 × 10−8) from this GWASs as genetic instruments (Table S1). Gut microbial metabolites Considering the important roles of gut microbiota- derived metabolites in microbiota-host crosstalk in the brain function and behavior, we further chose key me- tabolites with available GWAS, including propionic acid, β-hydroxybutyric acid (BHB), serotonin, GABA, tri-
  • 53. methylamine N-oxide (TMAO), betaine, choline, and carnitine. These gut microbial metabolites play crucial roles in maintaining a healthy neuropsychiatric function, and if dysregulated, potentially causally linked to neuro- psychiatric disorders according to previous studies [3, 24, 25]. We searched PubMed for GWASs of the gut metabolites and leveraged summary-level data from a re- cent GWAS of the human metabolome conducted among 2076 participants of the Framingham Heart Study (Table 1) [20]. Since few loci identified by gut me- tabolite GWAS have reached the level of genome-wide significance, we only selected SNPs at thresholds for suggestive genome-wide significance (P < 1 × 10−5) from the GWAS for each metabolite (Table S2). Neuropsychiatric disorders We searched PubMed for GWASs of the neuropsychi- atric disorders and identified SNPs with genome-wide significant (P < 5 × 10−8) associations for AD [21], MDD [22], and SCZ [23], respectively (Table 1, Table S3). Summarized data for AD were obtained from the Inter- national Genomics of Alzheimer’s Project (IGAP), in- cluding 25,580 AD cases and 48,466 controls, and the analysis was adjusted for age, sex, and principal compo- nents when necessary [21]. Genetic associations for MDD were obtained from Psychiatric Genomics Consor- tium 29 (PGC29) including135,458 MDD cases and 344, 901 controls, using sex and age as covariates [22]. Gen- etic associations for SCZ were obtained from a meta- analysis of Sweden and PGC including 13,833 SCZ cases and 18,310 controls [23]. Detailed information on diag- nostic criteria for AD, MDD, and SCZ are provided in Table S4. These GWASs identified 19 SNPs for AD, 44 SNPs for MDD, and 24 SNPs for SCZ (P < 5 × 10−8), re- spectively (Table S3).
  • 54. Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 2 of 9 Statistical analysis For instrumental variables, we only selected independent genetic variants which are not in linkage disequilibrium (LD) (defined as r2 < 0.1) with other genetic variants based on European ancestry reference data from the 1000 Genomes Project. We chose the variant with the lowest P value for association with the exposure when genetic variants were in LD. Moreover, for SNPs that were not available in GWASs of the outcome, we used the LD proxy search on the online platform (https:// snipa.helmholtz-muenchen.de/snipa3/index.php/) to re- place them with the proxy SNPs identified in high-LD (r2 > 0.8) or discard them if the proxies were not avail - able. Power calculations for the MR study were con- ducted based on the website: mRnd (http://cnsgenomics. com/shiny/mRnd/). We combined MR estimates by using inverse variance weighting (IVW) as primary method. Weighted mode, weighted median, and MR-Egger methods were used as sensitivity analyses. Detailed information about the MR methods mentioned above has been explained previously [26, 27]. The MR-Egger method examined for unknown horizontal pleiotropy as indicated by a non-zero inter- cept value. We also applied leave-one-SNP-out approach assessing the effects of removing these SNPs from the MR analysis to rule out potential pleiotropic effects. Ef- fect estimates are reported in beta values for the con- tinuous outcome and ORs (95% CIs) for binary outcome. Bonferroni correction was used to adjust for multiple comparisons, giving a cutoff of P = 7.6 × 10−4
  • 55. for the causal effect of gut microbiota on disorders and a cutoff of P = 1.7 × 10−4 for reverse causation. The MR analyses were conducted in the R version 3.5.1 computing environment (http://www.r-project.org) using the TwoSampleMR package (R project for Statis- tical Computing). This package harmonized effect of the exposure and outcome data sets including combined in- formation on SNPs, including phenotypes, effect alleles, effect allele frequencies, effect sizes, and standard errors for each SNP. In addition, we assumed that all alleles are presented on the forward strand in harmonization. In conclusion, the bi-directional MR results using the full set of selected SNPs. Results Associations of gut microbiota and metabolites with neuropsychiatric disorders We found suggestive evidence of a protective effect of the host-genetic-driven increase in Blautia on the risk of AD (per relative abundance: OR, 0.88; 95% CI, 0.79– 0.99; P = 0.028) (Fig. 1, Figure S1). Importantly, we fur- ther observed suggestive evidence that genetically ele- vated gut metabolite GABA was associated with a lower risk of AD (per 10 units: 0.96; 0.92–1.00; P = 0.034) (Figs. 1 and 2). Furthermore, the host-genetic-driven increases in En- terobacteriaceae family and Enterobacteriales order were potentially related to a higher risk of SCZ (1.09; 1.00– 1.18; P = 0.048), while Gammaproteobacteria class was related to a lower risk of SCZ (0.90; 0.83–0.98; P = 0.011) (Fig. 1, Figure S1). Interestingly, gut production of serotonin was potentially associated with a higher risk of
  • 56. SCZ (1.07; 1.00–1.15; P = 0.047) (Figs. 1 and 3). In addition, we found suggestive association of the host- genetic-driven increase in Bacilli class with a higher risk of MDD (1.07; 1.02–1.12; P = 0.010) (Fig. 1, Figure S1). Sensitivity analysis yielded similar results for the causal effects of gut microbiota on neuropsychiatric disorders, and no horizontal pleiotropy or outliers were observed (Tables S5 and S6). No significant results were found for any of other selected gut microbiota or metabolites with neuro- psychiatric disorders (Table S7). MR power calculation showed strong power to detect significant (P < 7.6 × 10−4) causal effect (OR = 1.2) for most of gut microbiota with the risk of AD, MDD, and SCZ, respectively (Table S8). Associations of neuropsychiatric disorders with gut microbiota In the opposite direction, we applied the MR method to investigate the causal relationship of neuropsychiatric Table 1 Description of gut microbiota, metabolites, and neuropsychiatric disorders Traits Consortium or study Sample size Populations Journal Year Gut Gut microbiota PopGen/FoCus 1812 individuals European Nat Genet. 2016 Gut metabolites FHS 2076 individuals European Cell Metab. 2013 Neuropsychiatric disorders
  • 57. Alzheimer’s disease IGAPa 25,580 cases and 48,466 controls European Nat Genet. 2013 Major depression disorder PGC29/deCODE/GenScotland/GERA/iPSYCH/UK Biobank/23andMeD 135,458 cases and 344,901 controls European Nat Genet. 2018 Schizophrenia Sweden/PGC 21,246 cases and 38,072 controls European Nat Genet. 2013 FoCus Food-Chain Plus, GERA Genetic Epidemiology Research on Adult Health and Aging, PGC Psychiatric Genomics Consortium a IGAP includes the Alzheimer’s Disease Genetics Consortium (ADGC), the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium (CHARGE), the European Alzheimer’s disease Initiative (EADI), and the Genetic and Environmental Risk in Alzheimer’s disease consortium (GERAD) Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 3 of 9 https://snipa.helmholtz-muenchen.de/snipa3/index.php https://snipa.helmholtz-muenchen.de/snipa3/index.php http://cnsgenomics.com/shiny/mRnd/ http://cnsgenomics.com/shiny/mRnd/ http://www.r-project.org disorders with gut microbiota. We found a suggestive as- sociation of AD with lower relative abundance of Erysi - pelotrichaceae family, Erysipelotrichales order, and Erysipelotrichia class (per 1-unit odds ratio: Beta±SE, −
  • 58. 0.274 ± 0.090; P = 0.003) and higher relative abundance of unclassified Porphyromonadaceae (0.351 ± 0.170; P = 0.040) (Fig. 1, Table S9). Additionally, MDD was associ - ated with higher relative abundance of unclassified Clos- tridiales (0.577 ± 0.241; P = 0.017), OTU16802 Bacteroides (0.842 ± 0.386; P = 0.029), and unclassified Prevotellaceae (0.978 ± 0.464; P = 0.035) (Fig. 1, Table S9). We further identified that SCZ was nominally re- lated to 2 genera, including higher relative abundance of OTU10589 unclassified Enterobacteriaceae (0.457 ± 0.220; P = 0.037) and lower relative abundance of un- classified Erysipelotrichaceae (− 0.248 ± -0.019; P = 0.045) (Fig. 1, Table S9). Associations were almost consistent in sensitivity ana- lyses using the weighted mode and weighted median methods. The MR-Egger method showed directional pleiotropy in the analysis of association between MDD and OTU16802 Bacteroides (P = 0.022) but not in any other potential significant associations (Table S9). How- ever, we had limited power (all less than 50%) to test sig- nificant (P < 1.7 × 10−4) causal effect (Beta = 0.5) of the risk of AD, MDD, and SCZ on specific gut microbiota (data not shown), possibly due to small sample size of the gut microbiota GWAS. Discussion In this two-sample bi-directional MR study, we found suggestive evidence of causal relationships of Blautia with AD, of Enterobacteriaceae family, Enterobacteriales order, and Gammaproteobacteria class with SCZ, and of Bacilli class with MDD. More importantly, several neu- rotransmitters such as GABA and serotonin produced by gut microbiota were also potentially linked to the risks of neuropsychiatric disorders, implying their im-
  • 59. portant roles in microbiota-host crosstalk in the brain function and behavior. In the other direction, our results suggested that neuropsychiatric disorders, including AD, SCZ, and MDD might alter the composition of gut microbiota. Microbiota-gut-brain communication has been shown to play a key role in cognitive function [2]. However, animal studies regarding the effects of Blautia genus on AD have yielded conflicting results, but extrapolating these findings to human beings is challenging [28, 29]. A cohort study (n = 108) reported that decreased propor- tion of Blautia hansenii was associated with a higher risk of AD [30], while two case-control studies observed that Blautia were more abundant in AD patients [5, 31]. Fig. 1 Schematic representation of the present study, highlighting for each step of the study design and the significant results obtained. We aimed to estimate causal relationships between gut microbiota (98 individual bacterial traits) and neuropsychiatric disorders (Alzheimer’s disease, major depression disorder, and schizophrenia) using a bi- directional Mendelian randomization (MR) approach (step 1). Then, we performed a two- sample MR analysis to identify which microbiota metabolites associated with these disorders (step 2). Finally, we identified 14 individual bacterial traits and 2 gut metabolites to be associated with these disorders. GABA, γ-aminobutyric acid; SCFA, short-chain fatty acids Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 4 of 9
  • 60. Although the direction of associations between Blautia and the risk of AD substantially differed across studies, one consistent finding was that gut microbial neuro- transmitter GABA, a downstream product of Blautia- dependent arginine metabolism, was related to a reduced risk of AD. Notably, lower levels of gut product of GABA were observed in patients with AD in several case-control studies [32, 33]. In this bi-directional MR study, our results for the first time provide evidence of a causal relationship between relative abundance of Blau- tia and AD. More importantly, we demonstrated that el- evated GABA was potentially associated with a lower risk of AD. Our findings supported previous meta- analysis of 35 observational studies which suggested that GABA level in AD were significantly lower than that of controls [12]. Our findings suggest that GABA produced by gut microbiota may play an important role in microbiota-host crosstalk in the brain function and be- havior. Although not significant, our findings show very similar association directions for Blautia with MDD and SCZ. Our findings are in line with recent studies which indicated that decreased Blautia was associated with an increased risk of autistic spectrum disorder (ASD), sug- gesting a general change associated with psychiatric dis- orders [34]. There are many potential pathways linking specific gut microbiota to AD, among which metabolites produced by gut microbiota may play an important role. It is worth noting that GABA, as a primary inhibitory neuro- transmitter in the human central nervous system (CNS), has been shown to shape neurological processes and cognition [35]. Recent evidence has demonstrated that GABAergic functions could be an essential factor in the whole stage of AD pathogenesis which seemed to be
  • 61. more resistant to neurodegenerative changes in aged brain [36, 37]. Our MR results that increased GABA levels was potentially associated with a lower risk of AD lent further support to the hypotheses. The biological mechanisms of GABA production include degradation of putrescine, decarboxylation of glutamate, or from ar- ginine or ornithine [8]. Interestingly, the genus Blautia has shown a strong correlation with arginine metabolism [38], which may be involved in AD pathogenesis by regulating its downstream products such as GABA, sup- porting the potential pathway [39]. Since AD does not break out suddenly but develops through a long pro- dromal phase instead, it is plausible that our findings may be potentially effective in early interventions of such dis - order in the future by targeting the microbiota (e.g., gut microbiota transplantation, psychobiotics, or antibiotics). Fig. 2 Causal effect of GABA with the risk of AD. a Schematic representation of the MR analysis results: genetically determined higher GABA plasma levels were potentially associated with a lower risk of AD. b The odds ratios (95% confidence interval) for AD per 10 units increase in GABA, as estimated in the inverse-variance weighted, weighted mode, weighted median, and MR-Egger MR analysis. The intercept of MR-Egger can be interpreted as a test of overall unbalanced horizontal pleiotropy. c The scatter plot represents instruments association including AD associations (y-axis) against instrument GABA associations (x- axis). The tunnel plot represents instrument precision (i.e., instrument AD regression coefficients divided by the correspondent instrument GABA SEs) (y-axis) against individual instrument ratio estimates in log odds ratio of AD (x- axis). βIV indicates odds ratio estimate per 1-ln 10 units
  • 62. increment in GABA levels. AD, Alzheimer’s disease; OR, odds ratio; CI, confidence interval; SNP, single-nucleotide polymorphism; SE, standard error; IVW, inverse variance weighted Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 5 of 9 Recently, Enterobacteriales family and Gammaproteo- bacteria class have been identified to be important bio- markers of SCZ in recent cross-sectional studies, consistent with our findings [6, 40]. Furthermore, a case- control study (n = 364) identified a strong relationship of lower platelet serotonin concentrations with depres- sive symptoms of SCZ [14]. However, available evidence is still largely inadequate since observational studies mainly rely on self-reported information and are suscep- tible to confounding (e.g., diet and health status) and re- verse causation bias. Ertugrul et al. observed plasma serotonin increased while platelet serotonin decreased in SCZ patients after clinical treatments, which was incon- sistent with our findings [13]. In addition, our results support the finding that increased Bacilli is potentially associated with a higher risk of MDD, possibly involving dopamine metabolism which might play a role in the major symptoms of MDD [41, 42]. A meta-analysis of RCTs showed that probiotics, typically including Lacto- bacillus and Bifidobacterium, had some benefit for MDD, but we found no associations for these micro- biota, possibly due to the synergistic effect of gut micro- biome so that the influence of a particular taxon may be different from multiple taxa [43]. Furthermore, these clinical trials might draw biased conclusions because of small sample sizes (ranging from 17 to 110) or short-
  • 63. term effects (ranging from 3 to 24 weeks). Therefore, a large and long-term RCT in a well-characterized popula- tion using probiotic capsules containing specific micro- biota might provide further evidence for the gut-brain axis in these disorders. Importantly, epidemiological study indicated that elevated Enterobacteriales was also associated with a higher risk of ASD, suggesting that the same changes in intestinal microbiota composition might lead to different outcomes due to gene-gene inter- actions and gene-environment interactions [44]. Al- though our results showed no significant association for Gammaproteobacteria and MDD, animal models found increased levels of Gammaproteobacteria were also asso- ciated with higher MDD risk and fluoxetine treatment was effective, implying strong correlations between gut microbiota and anxiety- and depression-like behaviors [45]. The serotonin hypothesis of SCZ originated from earl- ier studies of interactions between the hallucinogenic drug D-lysergic acid diethylamide and serotonin in per- ipheral systems. However, direct evidence of serotoner- gic dysfunction in the pathogenesis of SCZ remains unclear [46]. According to the principle of brain plasti - city, glutamate signals are destroyed by serotonergic overdrive, leading to neuronal hypometabolism, synaptic atrophy, and gray matter loss in the end [47]. Our find- ings that genetically increased serotonin levels was po- tentially related to a high risk of SCZ using a MR Fig. 3 Causal effect of serotonin with the risk of SCZ. a Schematic representation of the MR analysis results: genetically determined higher serotonin plasma levels were potentially associated with a higher risk of SCZ. b The odds ratios (95% confidence interval)
  • 64. for SCZ per 10 units increase in serotonin, as estimated in the inverse-variance weighted, weighted mode, weighted median, and MR-Egger MR analysis. The intercept of MR-Egger can be interpreted as a test of overall unbalanced horizontal pleiotropy. c The scatter plot represents instruments association including SCZ associations (y-axis) against instrument serotonin associations (x-axis). The tunnel plot represents instrument precision (i.e., instrument SCZ regression coefficients divided by the correspondent instrument serotonin SEs) (y-axis) against individual instrument ratio estimates in log odds ratio of SCZ (x-axis). βIV indicates odds ratio estimate per 1-ln 10 units increment in serotonin levels. SCZ, schizophrenia Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 6 of 9 approach supported such hypothesis. Importantly, En- terobacteriaceae family and Enterobacteriales order can produce SCFAs (e.g., acetic acid and formic acid) in carbohydrate fermentation, thus inducing serotonin bio- synthesis by enterochromaffin cells which are the major producers of serotonin, and ultimately increasing the risk of SCZ [48, 49]. Our novel findings highlighted the potentially important role of gut microbiota-related neu- rotransmitters in effective and benign therapies of psy- chiatric disorders. Furthermore, we also found that neuropsychiatric disor- ders might alter the composition of gut microbiota. Our findings were consistent with a small case-control study (n
  • 65. = 50) suggesting that Erysipelotrichaceae family were all less abundant in patients with AD [5]. An observational study showed that Porphyromonadaceae were associated with poor cognitive performance, partly consistent with our results [50]. However, the results from animal studies are conflicting. Although several animal studies suggested that anti-AD microbes, such as Erysipelotrichiaceae, decreased in mouse models with AD, and Porphyromonadaceae in- creased in aged mice [28, 51], other animal studies showed that the relative abundance of Erysipelotrichiaceae was positively correlated with AD [52, 53]. Therefore, the asso- ciation of neuropsychiatric disorders with specific gut microbiota requires further study. It is universally accepted that the CNS modulates gut microbiota compositions mainly through hypothalamic-pituitary-adrenal (HPA) axis, or classical neurotransmitters liberated by neuronal efferent activation, which explains the microbiota-host crosstalk in neuropsychiatric disorders from another direction [54]. Additionally, it is plausible that alterations in gut microbiota and related metabolites would lead to a sys- temic change in inflammation that may contribute to the neuroinflammation in AD, MDD, and SCZ. Increas- ing evidence suggests that bacteria populating the gut microbiome may excrete large quantities of lipopolysac- charides and amyloids, resulting in the pathogenesis of AD during aging when the permeability of gastrointes- tinal tract epithelium or blood-brain barrier increases [55]. Recent research has indicated that gut inflamma- tion can induce activation of microglia and the kynure- nine pathway, which activate systemic inflammation- inducing depressive or schizophrenic symptoms [56, 57]. Therefore, more studies are required to explore the mechanisms underlying the relationships of inflamma- tion with the gut microbiota-brain axis and its relations with AD, MDD and SCZ.
  • 66. Strengths of the present study included the bi-directional MR design and the use of summary-level data from thus far the largest GWASs. This design generally avoided bias due to reverse causation and confounding to obtain accurate results under MR assumptions. In addition, consistent re- sults from several sensitivity analyses including the use of weighted mode, weighted median, and MR-egger methods indicate robustness of our findings. However, several limita- tions merit consideration. First, our results did not survive a strict Bonferroni correction adjusting for multiple com- parisons, whereas as a hypothesis-driven approach, the MR study with some biological evidence was used to test epide- miologically established associations, regardless of Bonfer - roni corrected P values. Second, we used limited number of gut microbiota SNPs as instrumental variables; we cannot exclude that our findings might have been affected by weak instrument bias, although all genetic instruments were as- sociated with the exposure (F-statistic > 10). Third, statis- tical power was limited for associations of neuropsychiatric disorders with gut microbiota, so we cannot exclude type II error as an explanation for the null results completely. Lar - ger GWASs of gut microbiota are required to provide suffi - cient statistical power. However, the power was strong enough for the effect of gut microbiota on these disorders, which was our main findings in the present study. Fourth, our results were restricted to European ancestry. Replica- tion with functionally relevant genetic prediction of gut microbiota is warranted given the substantial difference in gut microbiota composition among different populations. Fifth, the 16S rRNA gene sequencing only permit reso- lution from the genus to the phylum level rather than at a more specific level, resulting in biased results if some spe- cific species contributed to neuropsychiatric disorders. Fi - nally, gut microbiota might be influenced by environmental
  • 67. factors such as dietary habits or health status, which led to lower variance explained by genetic instruments. However, we could not test whether genetic instruments are associ- ated with these confounders such as diet or lifestyle infor - mation in the present study where such information is not available. Conclusions In summary, our findings supported several potential as- sociations between specific gut microbiota and neuro- psychiatric disorders and highlighted the important roles of microbial neurotransmitters such as GABA and sero- tonin in microbiota-host crosstalk in neuropsychiatric disorders. Further investigations in understanding the underlying mechanisms of gut microbiota in the devel- opment of neuropsychiatric disorders are required. Supplementary information Supplementary information accompanies this paper at https://doi.org/10. 1186/s12974-020-01961-8. Additional file 1: Figure S1. Odds ratio for association of genetically predicted gut microbiota with neuropsychological diseases. Table S1. Characteristics of selected SNPs for core gut microbiota. Table S2. Characteristics of selected SNPs for gut metabolites. Table S3. Characteristics of selected SNPs for neuropsychological diseases. Table S4. Description of the diagnostic assessment for neuropsychological Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 7 of 9
  • 68. https://doi.org/10.1186/s12974-020-01961-8 https://doi.org/10.1186/s12974-020-01961-8 diseases. Table S5. Associations between genetically predicted gut microbiota and neuropsychological diseases in sensitivity analyses. Table S6. Associations between genetically predicted gut microbiota and neuropsychological diseases in a leave-one-out approach. Table S7. As- sociations between genetically predicted metabolites and neuropsycho- logical diseases using IVW method. Table S8. MR Power calculation for detecting significant (P < 7.6 × 10-4) causal effect (OR = 1.2) of gut micro- biome on the risk of AD, MDD, and SCZ. Table S9. Effect estimates for association of genetically predicted neuropsychological diseases with gut microbiota using four Mendelian randomization methods. Abbreviations AD: Alzheimer’s disease; MDD: Major depressive disorder; SCZ: Schizophrenia; MR: Mendelian randomization; GWAS: Genome-wide association study; GABA: γ-Aminobutyric acid; SCFA: Short-chain fatty acid; BHB: β- Hydroxybutyric acid; TMAO: Trimethylamine N-oxide; SNP: Single nucleotide polymorphism; IGAP: International Genomics of Alzheimer’s Project;
  • 69. PGC29: Psychiatric Genomics Consortium 29; LD: Linkage disequilibrium; IVW: Inverse variance weighting; CNS: Central nervous system; HPA: Hypothalamic-pituitary-adrenal; ASD: Autistic spectrum disorder Acknowledgements The PopGen 2.0 network (P2N) is supported by a grant from the German Federal Ministry for Education and Research (01EY1103). We thank Drs. Andre Franke and Wolfgang Lieb for sharing the GWAS summary data for beta diversity and bacterial abundance from published paper (Nat Genet. 2016 Nov; 48(11): 1396-1406.) Authors’ contributions ZZ, LQ, and TH designed the research. ZZ and TH had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. ZZ, LQ, and TH wrote the paper and performed the data analysis. All authors contributed to the statistical analysis, critically reviewed the manuscript during the writing process, and approved the final version to be published. ZZ and TH are the guarantors for the study. Funding The study was supported by grants from the National Key Research and Development Project (2019YFC2003400), the Peking University
  • 70. Start-up Grant (BMU2018YJ002), High-performance Computing Platform of Peking Univer- sity, and the China-Canada Key Lab of Nutrition and Health at Beijing Tech- nology and Business University- Grant: 88442Y0033. The funding organization had no role in the preparation of the manuscript. Availability of data and materials All data used in the present study were obtained from genome- wide associ- ation study summary statistics which were publicly released by genetic consortia. Ethics approval and consent to participate Contributing studies received ethical approval from their respective institutional review boards. Consent for publication Not applicable. Competing interests All authors declare no support from companies for the submitted work; no relationships with companies that might have an interest in the submitted work in the previous 3 years; no spouses, partners, or children that have financial relationships that may be relevant to the submitted work; and no non-financial interests that may be relevant to the submitted work.
  • 71. Author details 1Department of Epidemiology & Biostatistics, School of Public Health, Peking University, 38 Xueyuan Road, Beijing 100191, China. 2Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA. 3Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA. 4Department of Global Health, School of Public Health, Peking University, Beijing 100191, China. 5Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing 100191, China. 6Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing 100191, China. Received: 2 July 2020 Accepted: 23 September 2020 References 1. Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang J, Tito RY, Schiweck C, Kurilshikov A, Joossens M, Wijmenga C, et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. 2019;4(4):623–32. 2. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat Neurosci.
  • 72. 2017;20(2):145–55. 3. Caspani G, Swann J. Small talk: microbial metabolites involved in the signaling from microbiota to brain. Curr Opin Pharmacol. 2019;48:99–106. 4. van de Wouw M, Boehme M, Lyte JM, Wiley N, Strain C, O'Sullivan O, Clarke G, Stanton C, Dinan TG, Cryan JF. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain-gut axis alterations. J Physiol. 2018;596(20):4923–44. 5. Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg H, Blennow K, et al. Gut microbiome alterations in Alzheimer’s disease. Sci Rep. 2017;7(1):13537. 6. Shen Y, Xu J, Li Z, Huang Y, Yuan Y, Wang J, Zhang M, Hu S, Liang Y. Analysis of gut microbiota diversity and auxiliary diagnosis as a biomarker in patients with schizophrenia: a cross-sectional study. Schizophr Res. 2018;197: 470–7. 7. Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linlokken A, Wilson R, Rudi K. Correlation between the human fecal microbiota and depression. Neurogastroenterol Motil. 2014;26(8):1155–62. 8. Strandwitz P, Kim KH, Terekhova D, Liu JK, Sharma A,
  • 73. Levering J, McDonald D, Dietrich D, Ramadhar TR, Lekbua A, et al. GABA- modulating bacteria of the human gut microbiota. Nat Microbiol. 2019;4(3):396–403. 9. Strandwitz P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018;1693(Pt B):128–33. 10. Huang D, Liu D, Yin J, Qian T, Shrestha S, Ni H. Glutamate-glutamine and GABA in brain of normal aged and patients with cognitive impairment. Eur Radiol. 2017;27(7):2698–705. 11. Peitl V, Štefanović M, Karlović D. Depressive symptoms in schizophrenia and dopamine and serotonin gene polymorphisms. Prog Neuropsychopharmacol Biol Psychiatry. 2017;77:209–15. 12. Manyevitch R, Protas M, Scarpiello S, Deliso M, Bass B, Nanajian A, Chang M, Thompson SM, Khoury N, Gonnella R, et al. Evaluation of metabolic and synaptic dysfunction hypotheses of Alzheimer’s disease (AD): a meta- analysis of CSF markers. Curr Alzheimer Res. 2018;15(2):164– 81. 13. Ertugrul A, Ucar G, Basar K, Demir B, Yabanoglu S, Ulug B. Influence of clozapine on platelet serotonin, monoamine oxidase and plasma serotonin levels. Psychiatry Res. 2007;149(1-3):49–57. 14. Peitl V, Vidrih B, Karlović Z, Getaldić B, Peitl M, Karlović
  • 74. D. Platelet serotonin concentration and depressive symptoms in patients with schizophrenia. Psychiatry Res. 2016;239:105–10. 15. Emdin CA, Khera AV, Kathiresan S. Mendelian randomization. JAMA. 2017; 318(19):1925–6. 16. Sanna S, van Zuydam NR, Mahajan A, Kurilshikov A, Vich Vila A, Võsa U, Mujagic Z, Masclee AAM, Jonkers DMAE, Oosting M, et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet. 2019;51(4):600–5. 17. Turpin W, Espin-Garcia O, Xu W, Silverberg MS, Kevans D, Smith MI, Guttman DS, Griffiths A, Panaccione R, Otley A, et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat Genet. 2016;48(11):1413–7. 18. Goodrich JK, Davenport ER, Beaumont M, Jackson MA, Knight R, Ober C, Spector TD, Bell JT, Clark AG, Ley RE. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe. 2016;19(5):731– 43. 19. Wang J, Thingholm LB, Skiecevičienė J, Rausch P, Kummen M, Hov JR, Degenhardt F, Heinsen F-A, Rühlemann MC, Szymczak S, et al. Genome-
  • 75. wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat Genet. 2016;48(11):1396– 406. 20. Rhee EP, Ho JE, Chen MH, Shen D, Cheng S, Larson MG, Ghorbani A, Shi X, Helenius IT, O'Donnell CJ, et al. A genome-wide association study of the Zhuang et al. Journal of Neuroinfla mmation (2020) 17:288 Page 8 of 9 human metabolome in a community-based cohort. Cell Metab. 2013;18(1): 130–43. 21. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, DeStafano AL, Bis JC, Beecham GW, Grenier-Boley B, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45(12):1452–8. 22. Wray NR, Ripke S, Mattheisen M, Trzaskowski M, Byrne EM, Abdellaoui A, Adams MJ, Agerbo E, Air TM, Andlauer TMF, et al. Genome- wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat Genet. 2018;50(5):668–81.
  • 76. 23. Ripke S, O'Dushlaine C, Chambert K, Moran JL, Kähler AK, Akterin S, Bergen SE, Collins AL, Crowley JJ, Fromer M, et al. Genome-wide association analysis identifies 13 new risk loci for schizophrenia. Nat Genet. 2013;45(10): 1150–9. 24. Sabokdast M, Habibi-Rezaei M, Moosavi-Movahedi AA, Ferdousi M, Azimzadeh-Irani E, Poursasan N. Protection by beta- Hydroxybutyric acid against insulin glycation, lipid peroxidation and microglial cell apoptosis. Daru. 2015;23:42. 25. Zalar B, Haslberger A, Peterlin B. The role of microbiota in depression - a brief review. Psychiatr Danub. 2018;30(2):136–41. 26. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40(4):304–14. 27. Hartwig FP, Davey Smith G, Bowden J. Robust inference in summary data Mendelian randomization via the zero modal pleiotropy assumption. Int J Epidemiol. 2017;46(6):1985–98. 28. Xin Y, Diling C, Jian Y, Ting L, Guoyan H, Hualun L, Xiaocui T, Guoxiao L, Ou S, Chaoqun Z, et al. Effects of oligosaccharides from Morinda officinalis on
  • 77. gut microbiota and metabolome of APP/PS1 transgenic mice. Front Neurol. 2018;9:412. 29. Park J-Y, Choi J, Lee Y, Lee J-E, Lee E-H, Kwon H-J, Yang J, Jeong B-R, Kim Y- K, Han P-L. Metagenome analysis of bodily microbiota in a mouse model of Alzheimer disease using bacteria-derived membrane vesicles in blood. Exp Neurobiol. 2017;26(6):369–79. 30. Haran JP, Bhattarai SK, Foley SE, Dutta P, Ward DV, Bucci V, McCormick BA. Alzheimer’s disease microbiome is associated with dysregulation of the anti- inflammatory P-glycoprotein pathway. mBio. 2019;10(3):e00632–19. 31. Li B, He Y, Ma J, Huang P, Du J, Cao L, Wang Y, Xiao Q, Tang H, Chen S. Mild cognitive impairment has similar alterations as Alzheimer’s disease in gut microbiota. Alzheimers Dement. 2019;15(10):1357–66. 32. Gueli MC, Taibi G. Alzheimer’s disease: amino acid levels and brain metabolic status. Neurol Sci. 2013;34(9):1575–9. 33. Jiménez-Jiménez FJ, Molina JA, Gómez P, Vargas C, de Bustos F, Benito- León J, Tallón-Barranco A, Ortí-Pareja M, Gasalla T, Arenas J. Neurotransmitter amino acids in cerebrospinal fluid of patients with Alzheimer’s disease. J Neural Transm (Vienna). 1998;105(2- 3):269–77.
  • 78. 34. Liu F, Li J, Wu F, Zheng H, Peng Q, Zhou H. Altered composition and function of intestinal microbiota in autism spectrum disorders: a systematic review. Transl Psychiatry. 2019;9(1):43. 35. de JR De-Paula V, Forlenza AS, Forlenza OV. Relevance of gutmicrobiota in cognition, behaviour and Alzheimer’s disease. Pharmacol Res. 2018;136:29–34. 36. Rissman RA, De Blas AL, Armstrong DM. GABA(A) receptors in aging and Alzheimer’s disease. J Neurochem. 2007;103(4):1285–92. 37. Govindpani K, Calvo-Flores Guzmán B, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Towards a better understanding of GABAergic remodeling in Alzheimer’s disease. Int J Mol Sci. 2017;18(8):1813. 38. Liu X, Zheng H, Lu R, Huang H, Zhu H, Yin C, Mo Y, Wu J, Liu X, Deng M, et al. Intervening effects of total alkaloids of Corydalis saxicola Bunting on rats with antibiotic-induced gut microbiota dysbiosis based on 16S rRNA gene sequencing and untargeted metabolomics analyses. Front Microbiol. 2019;10:1151. 39. Bergin DH, Jing Y, Zhang H, Liu P. A single intracerebroventricular Aβ25-35 infusion leads to prolonged alterations in arginine metabolism in the rat
  • 79. hippocampus and prefrontal cortex. Neuroscience. 2015;298:367–79. 40. Nguyen TT, Kosciolek T, Maldonado Y, Daly RE, Martin AS, McDonald D, Knight R, Jeste DV. Differences in gut microbiome composition between persons with chronic schizophrenia and healthy comparison subjects. Schizophr Res. 2019;204:23–9. 41. Cao G, Tao F, Hu Y, Li Z, Zhang Y, Deng B, Zhan X. Positive effects of a Clostridium butyricum-based compound probiotic on growth performance, immune responses, intestinal morphology, hypothalamic neurotransmitters, and colonic microbiota in weaned piglets. Food Funct. 2019;10(5):2926–34. 42. Opmeer EM, Kortekaas R, Aleman A. Depression and the role of genes involved in dopamine metabolism and signalling. Prog Neurobiol. 2010; 92(2):112–33. 43. Barbosa RSD, Vieira-Coelho MA. Probiotics and prebiotics: focus on psychiatric disorders - a systematic review. Nutr Rev. 2020;78(6):437-50. 44. Liu S, Li E, Sun Z, Fu D, Duan G, Jiang M, Yu Y, Mei L, Yang P, Tang Y, et al. Altered gut microbiota and short chain fatty acids in Chinese children with
  • 80. autism spectrum disorder. Sci Rep. 2019;9(1):287. 45. Sun L, Zhang H, Cao Y, Wang C, Zhao C, Wang H, Cui G, Wang M, Pan Y, Shi Y, et al. Fluoxetine ameliorates dysbiosis in a depression model induced by chronic unpredicted mild stress in mice. Int J Med Sci. 2019;16(9):1260–70. 46. Yang AC, Tsai S-J. New targets for schizophrenia treatment beyond the dopamine hypothesis. Int J Mol Sci. 2017;18(8):1689. 47. Abi-Dargham A, Laruelle M, Aghajanian GK, Charney D, Krystal J. The role of serotonin in the pathophysiology and treatment of schizophrenia. J Neuropsychiatry Clin Neurosci. 1997;9(1):1–17. 48. Ge X, Pan J, Liu Y, Wang H, Zhou W, Wang X. Intestinal crosstalk between microbiota and serotonin and its impact on gut motility. Curr Pharm Biotechnol. 2018;19(3):190–5. 49. Blander JM, Longman RS, Iliev ID, Sonnenberg GF, Artis D. Regulation of inflammation by microbiota interactions with the host. Nat Immunol. 2017; 18(8):851–60. 50. Bajaj JS, Ahluwalia V, Steinberg JL, Hobgood S, Boling PA, Godschalk M, Habib S, White MB, Fagan A, Gavis EA, et al. Elderly patients have an altered gut-brain axis regardless of the presence of cirrhosis. Sci Rep.
  • 81. 2016;6:38481. 51. Du C-T, Gao W, Ma K, Yu S-X, Li N, Yan S-Q, Zhou F-H, Liu Z-Z, Chen W, Lei L-C, et al. MicroRNA-146a deficiency protects against Listeria monocytogenes infection by modulating the gut microbiota. Int J Mol Sci. 2018;19(4):993. 52. Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G, Neher JJ, Fak F, Jucker M, Lasser T, et al. Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci Rep. 2017;7:41802. 53. Bauerl C, Collado MC, Diaz Cuevas A, Vina J, Perez Martinez G. Shifts in gut microbiota composition in an APP/PSS1 transgenic mouse model of Alzheimer’s disease during lifespan. Lett Appl Microbiol. 2018;66(6):464–71. 54. Montiel-Castro AJ, González-Cervantes RM, Bravo-Ruiseco G, Pacheco-López G. The microbiota-gut-brain axis: neurobehavioral correlates, health and sociality. Front Integr Neurosci. 2013;7:70. 55. Lin L, Zheng LJ, Zhang LJ. Neuroinflammation, gut microbiome, and Alzheimer’s disease. Mol Neurobiol. 2018;55(11):8243–50. 56. Simkin DR. Microbiome and mental health, specifically as it relates to
  • 82. adolescents. Curr Psychiatry Rep. 2019;21(9):93. 57. Inta D, Lang UE, Borgwardt S, Meyer-Lindenberg A, Gass P. Microglia activation and schizophrenia: lessons from the effects of minocycline on postnatal neurogenesis, neuronal survival and synaptic pruning. Schizophr Bull. 2017;43(3):493–6. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Zhuang et al. Journal of Neuroinflammation (2020) 17:288 Page 9 of 9 AbstractBackgroundMethodsResultsConclusionsBackgroundMet hodsStudy design overviewData sources and instrumentsGut microbiotaGut microbial metabolitesNeuropsychiatric disordersStatistical analysisResultsAssociations of gut microbiota and metabolites with neuropsychiatric disordersAssociations of neuropsychiatric disorders with gut microbiotaDiscussionConclusionsSupplementary informationAbbreviationsAcknowledgementsAuthors’ contributionsFundingAvailability of data and materialsEthics approval and consent to participateConsent for publicationCompeting interestsAuthor detailsReferencesPublisher’s Note OPEN ORIGINAL ARTICLE
  • 83. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition M-L Wong1,2,9,10, A Inserra1,2,9, MD Lewis1,2, CA Mastronardi3, L Leong4,5, J Choo4,5, S Kentish6, P Xie7,10, M Morrison8, SL Wesselingh4,5, GB Rogers4,5,10 and J Licinio1,2,10 The inflammasome is hypothesized to be a key mediator of the response to physiological and psychological stressors, and its dysregulation may be implicated in major depressive disorder. Inflammasome activation causes the maturation of caspase-1 and activation of interleukin (IL)-1β and IL-18, two proinflammatory cytokines involved in neuroimmunomodulation, neuroinflammation and neurodegeneration. In this study, C57BL/6 mice with genetic deficiency or pharmacological inhibition of caspase-1 were screened for anxiety- and depressive-like behaviors, and locomotion at baseline and after chronic stress. We found that genetic deficiency of caspase-1 decreased depressive- and anxiety-like behaviors, and conversely increased locomotor activity and skills. Caspase-1 deficiency also prevented the exacerbation of depressive-like behaviors following chronic stress. Furthermore, pharmacological caspase-1 antagonism with minocycline ameliorated stress-induced depressive-like behavior in wild- type mice. Interestingly, chronic stress or pharmacological inhibition of caspase-1 per se altered the fecal microbiome in a very similar manner. When stressed mice were treated with minocycline, the observed gut microbiota changes included increase in relative abundance of Akkermansia spp. and Blautia spp., which are compatible
  • 84. with beneficial effects of attenuated inflammation and rebalance of gut microbiota, respectively, and the increment in Lachnospiracea abundance was consistent with microbiota changes of caspase-1 deficiency. Our results suggest that the protective effect of caspase-1 inhibition involves the modulation of the relationship between stress and gut microbiota composition, and establishes the basis for a gut microbiota–inflammasome–brain axis, whereby the gut microbiota via inflammasome signaling modulate pathways that will alter brain function, and affect depressive- and anxiety-like behaviors. Our data also suggest that further elucidation of the gut microbiota–inflammasome–brain axis may offer novel therapeutic targets for psychiatric disorders. Molecular Psychiatry (2016) 21, 797–805; doi:10.1038/mp.2016.46; published online 19 April 2016 INTRODUCTION Increasing evidence suggests an involvement of neuroinflamma- tory pathways in the etiopathophysiology of major depressive disorder (MDD) and antidepressant response.1,2 Depressive symptoms are underlined by increased levels of proinflammatory cytokines (that is, interleukin (IL)-1β and IL-6), decreased levels of anti-inflammatory cytokines (that is, IL-4 and IL-10) and are associated with polymorphisms in inflammation-related genes.3–5 IL-1 receptor type-I and its ligands are expressed in brain areas relevant to stress response,6–8 and IL-1β signaling is fundamental in mediating the deleterious neurobehavioral and